Abstract
Unique compositional and functional features of the cervicovaginal microbiota have been associated with protection against and risk for sexually transmitted infections (STI). In men, our knowledge of the interaction between the penile microbiota and STI is less developed. The current state of our understanding of these microbiota and their role in select STIs is briefly reviewed, along with strategies that leverage existing findings to manipulate genital microbiota and optimize protection against STIs. Finally, we focus on major research gaps and present a framework for future studies.
Keywords: anaerobes, Lactobacillus, microbiome, penile, STI, vagina
Amid reports of steep and sustained increases in gonorrhea, chlamydia, and syphilis [1], continued high rates transmission of human immunodeficiency virus-1 (HIV) [2], and new concerns regarding the development of antibiotic-resistant gonorrhea [3], novel antibiotic-sparing strategies to prevent acquisition of sexually transmitted infections (STIs) are urgently needed. Together with mucosal immune mechanisms, the complex and dynamic communities of bacteria inhabiting the human genital tract, the genital microbiota, may comprise the first lines of defense against invading pathogens [4]. Research suggests that vaginal microbiota play a critical role in host protection and susceptibility to STIs (including HIV) [5–8] as well as in poor obstetric outcomes such as preterm birth and stillbirth [9–11]. Emerging data also suggest a role for the male genital microbiota in HIV and STI susceptibility [12–14]. While additional foundational research is still needed, in the long term, improving our understanding of the role of the genital microbiota and its relationship to STIs has the potential to lead to new prevention strategies and to novel methods to improve reproductive health in the populations disproportionately affected by these infections. In this article, we discuss the genital microbiota in cis-gender women and men, and briefly review epidemiologic links between the genital microbiota and acquisition of select STIs focusing on heterosexual intercourse, as well as potential mechanisms mediating these links. Finally, we present an extended discussion of research gaps and future directions, and propose a framework for further studies.
THE FEMALE AND MALE GENITAL MICROBIOTA
Anatomic and Immunologic Differences, Shared Exposures
The male and female genital tracts represent distinct, complex systems in which anatomic and immunologic factors, hormones, and the microbiota interact to affect STI susceptibility. Landmarks in the male genital tract include foreskin, glans, coronal sulcus, and penile shaft, while the female genital tract comprises cervix, fornices, and vaginal canal [15, 16]. Women are at increased risk of acquisition of many STIs through heterosexual sex as compared to men who report sex with women. Anatomic and immunologic differences likely impact these gender discrepancies. A complete description of these differences is outside the scope of this review [16]; however, for example, the surface area of the cervicovaginal mucosa is considerably larger than that of the penis and foreskin, resulting in greater potential exposure to STI pathogens. Semen may remain within the female genital tract for up to 3 days postcoitus, prolonging exposure to STIs, including HIV [17]. In addition, women have increased mucosal expression of the HIV coreceptor CCR5 in the genital tract as compared to men [17] and female sex hormones such as estrogen and progesterone (whether endogenous or exogenous, eg, from use of hormonal contraception) may also affect STI susceptibility [18]. For example, under the influence of progesterone, cervicovaginal mucus is thick and viscous, which helps to block the movement of viral particulates, including STIs, from the lower to upper female genital tract [19, 20]. In contrast, during ovulation, with increasing estradiol, the mucus thins and becomes less viscous [21]. In vitro studies have shown that the cervicovaginal microbiota can modulate the penetration of HIV through mucus to access target cells [20]. During sexual activity, an individual’s genital microbiota can become exposed to the partner’s oral, genital, and rectal microbiota. Despite these exposures, distinctive genital microbial communities are observed in men and women, a phenomenon that reflects that at least for opposite-sex interactions, strong selective forces are exerted by sex-specific microenvironments. Below we outline current knowledge regarding the composition of vaginal and penile microbiota and their determinants. Of note, the pharynx and rectum are also important sites for STI/HIV exposure and infection both in men and women. However, there is a paucity of data on the relationship between the microbiota of these extragenital sites and STI susceptibility [22], and they are outside the scope of this review.
THE FEMALE GENITAL (CERVICOVAGINAL) MICROBIOTA
Female Genital Microbiota Structure
The vaginal microbiota has been investigated for over 120 years, beginning with morphological descriptions via microscopy [23]. It is dominated in a healthy or optimal state, by specific Lactobacillus spp., which produce lactic acid and help to maintain a protective, low-pH vaginal microenvironment [24]. In some women, for reasons that are not fully understood, these lactobacilli are decreased and the vaginal microbiota is comprised predominantly of a spectrum of strict and facultative anaerobic bacteria [5, 25, 26]. These types of microbiota are common and consistent with those in women diagnosed clinically with bacterial vaginosis (BV). BV affects over 27% of women in the United States [26–28]. Prevalence ranges from 23% to 29% throughout the rest of the world with substantially higher rates (over 50%) seen in select populations [27, 28]. Approximately 50% of women with BV are symptomatic, generally complaining of vaginal odor and discharge [29]. Symptomatic BV causes significant morbidity: it is the leading cause of vaginal symptoms in women of reproductive age. BV is associated with an increased risk of several adverse reproductive health outcomes, including preterm birth, low birth weight, and upper reproductive tract infections [8–11], However, while current guidelines recommend treatment of BV only in women complaining of symptoms [30], both symptomatic and asymptomatic BV have been associated with poor outcomes, including increased acquisition and transmission of STIs and HIV [7, 8, 26, 31].
BV is diagnosed in the clinical setting using Amsel’s criteria defined by the presence of at least 3 of the following: thin homogenous vaginal discharge, vaginal pH > 4.5, positive “whiff” test with addition of KOH to vaginal secretions, and presence of clue cells on microscopy [26, 29, 32], also referred to as Amsel-BV [26]. Commercial molecular tests for BV are also increasingly available for clinical use [33]. In research settings, diagnosis has historically been based on the Nugent score, derived from a Gram stain of vaginal secretions (a high score of 7–10 represents BV, ie, Nugent-BV) [26, 32]. More recently, new molecular approaches have provided an in-depth understanding of the vaginal microbiota composition, enabling species-level identification of microorganisms [26, 34–46]; this has also translated into advanced diagnostic methods that comparatively quantify relevant species [47, 48]. Studies have shown that reproductive aged women can be grouped into several distinct vaginal microbial community state types (CSTs) (Figure 1) based on diversity and relative abundance of bacteria, several dominated by Lactobacillus spp. (CST I dominated by Lactobacillus crispatus, CST II with Lactobacillus gasseri, CST III with Lactobacillus iners, and CST V with Lactobacillus jensenii). Low-Lactobacillus communities (CST IV) comprise of a variety of anaerobic bacteria [44]. Low-Lactobacillus communities with abundant anaerobes characterized by molecular techniques can be collectively termed molecular-BV (as proposed in a recent consensus position paper) [26]. Importantly, while the vaginal microbiota in some women may remain stable over time, in others it can fluctuate dramatically, over hours to days [43, 49] (Figure 1).
Figure 1.
A, Studies have shown that reproductive-aged women can be grouped into several distinct vaginal microbial community state types (CSTs) based on diversity and relative abundance of bacteria [44, 50]. There are 4 species of Lactobacillus that are consistently observed in surveys of vaginal microbiota and each is capable of dominating a vaginal community state type. CST I is dominated by Lactobacillus crispatus, CST II by L. gasseri, CST III by L. iners, and CST V by L. jensenii. Low-Lactobacillus communities (CST IV-A, CST IV-B, and CST IV-C) are dominated by a variety of bacterial vaginosis-associated bacteria (BVAB; eg, Atopobium, Prevotella, Dialister, Gardnerella, Megasphaera, Peptoniphilus, Sneathia, Eggerthella, Aerococcus, Finegoldia, and Mobiluncus) [44]. These low-Lactobacillus communities (defined by tools such as 16S rRNA gene amplicon sequencing, quantitative PCR, and metagenomic sequencing) are collectively termed molecular-BV [26]. B, While the vaginal microbiota in some women may remain stable over time dominated by Lactobacillus spp. or in the absence of Lactobacillus, in others it can fluctuate dramatically, over hours to days [43, 49].
Determinants of Female Genital Microbiota Structure and Dynamics
A number of host and exogenous or behavioral factors, including sex hormones, pregnancy, race, condom use, male partner circumcision, menses, sexual activity, antibiotics, douching, lubricant use, and smoking likely impact the cervicovaginal microbiota, although many causal mechanisms remain unknown or incompletely understood (Table 1). The female sex hormone estrogen is thought to promote the production of glycogen by vaginal epithelial cells. Glycogen can be broken down into glucose and maltose by bacterial and human amylases to support the growth of lactobacilli and other bacteria [51, 52]. One can hypothesize that after lowering the pH, Lactobacillus species use amylases (which unlike amylases from other anaerobes such as Gardnerella vaginalis preferentially operate at low pH [52]) to degrade available glycogen, thus selecting out competition for this precious resource and favoring its growth. Importantly, changes in estrogen levels throughout the life cycle are mirrored by corresponding changes in the vaginal microbiota [53] (Figure 2).
Table 1.
Factors Associated With Female Genital Microbiota Structure and Stability
Increased Risk of BV/Vaginal Microbiota Instability | Decreased Risk of BV/Vaginal Microbiota Instability |
---|---|
Menses | Estrogen |
New sexual partner | Hormonal contraception |
Condomless vaginal sex | Circumcised male partner |
Uncircumcised male partner | Pregnancy |
Female partner with BV | Antibiotics |
Black race | |
Smoking | |
Lubricant use | |
Douching |
Abbreviation: BV, bacterial vaginosis.
Figure 2.
A, Although Lactobacillus spp. may sometimes be found, the vaginal microbiota of premenarchal girls is generally characterized by low quantities of these bacteria. They are often present after birth, in part likely due to maternal estrogen stimulating the temporary maturation of the vaginal epithelium that supports the growth of Lactobacillus spp. As girls progress through puberty and eventually menarche (and estrogen levels increase [54]) the vaginal microbiota can transition to become Lactobacillus dominated [55, 56]. In menopause, as estrogen levels decline, Lactobacillus spp. decrease [57–61]. Pregnancy, a high-estrogen and -progesterone state, is associated with increased stability and Lactobacillus dominance of the vaginal microbiota [62, 63]. B, Increased estrogen at certain times of the menstrual cycle may correlate with increased Lactobacilli and increased stability [43]. Finally, exogenous sex hormones in the form of hormonal contraceptives, or topical estrogens used to treat atrophic vaginitis in postmenopausal women, have also been shown in some [64, 65], although not all studies [34] to increase vaginal Lactobacillus. The effects of environmental mimetic estrogens or xenoestrogens on the vaginal microbiota are currently unknown [66].
In North American populations, low-Lactobacillus cervicovaginal microbiota are more common among African American and Hispanic women compared to white women [44, 67, 68]. Indeed, nearly 50% of African American women may have low-Lactobacillus cervicovaginal microbiota [69]. The reasons for this high prevalence remain unknown; however, it has been postulated that psychosocial stress, sexual networks, diet, body mass index, male partner characteristics, or socioeconomic status might play a role [55, 70–75]. It is also possible that biological or genetic factors may mediate this association [27].
Menses has been shown to be associated with major changes of the vaginal microbiota and overgrowth of anaerobic bacteria (Figure 2), even though L. iners has been reported to dominate during menses in some women otherwise dominated by L. crispatus outside of menses [43, 49]. Multiple factors including blood introduced into the vaginal vault (different physical and biochemical environment), hormonal shifts, and behavioral changes (eg, the use of tampons or pads) may contribute to these shifts, which appear to be highly personalized [43, 49]. Male circumcision has been shown in a large randomized controlled trial to decrease BV in female partners [76]. Vaginal intercourse, especially condomless sex, may destabilize Lactobacillus-dominated vaginal microbiota [43], causing a shift to a predominance of anaerobic bacteria [77]. Douching and lubricant use are each associated with the onset of BV [78, 79]. Even more efficient sexual transmission of BV-associated bacteria may occur between female sex partners, who are typically concordant for the presence of BV and in whom BV is associated with sexual behaviors likely to transmit vaginal fluid [80–84]. Finally, antibiotic treatment whether for BV or other conditions may affect the vaginal microbiota [49, 85, 86].
THE MALE GENITAL MICROBIOTA
Structure and Composition of Male Genital Microbiota
The male genital microbiota has been much less studied than the cervicovaginal microbiota. Most existing studies have utilized swabs taken from the coronal sulcus (reflective of the penile microbiota), although a few have also utilized urine samples, which may be more reflective of the urethral microbiota. Studies utilizing swabs taken from the coronal sulcus demonstrate that the penile microbiota commonly contains bacteria similar to those found on the skin including Corynebacterium and Staphylococcus spp., as well as Anerococcus [87, 88]. Furthermore, the penile microbiota may contain anaerobic bacteria, eg, Clostridiales, Porphyromonas, and many anaerobic bacteria commonly associated with BV, such as Prevotella [87, 89]. A study of 165 uncircumcised participants grouped the penile microbiota into 7 distinct penile CSTs, defined both by bacterial composition and absolute abundance, wherein CSTs 1–3 contained taxa such as Corynebacterium and Lactobacillus, and CSTs 4–7 had relatively greater diversity and abundances of BV-associated anaerobic bacteria, including Prevotella and Porphyromonas, and Clostridiales [90]. While the corona sulcus might be important to HIV infection, STIs such as gonorrhea and chlamydia cause urethritis, thus the urethral microbiota might be more relevant to these infections. Significant differences were observed between the urethral (using urine as a surrogate) and corona sulcus microbiota [87]. Specifically, bacterial taxa such as Atopobium, Megasphaera, Mobiluncus, Prevotella, and Gemella were detected in coronal sulcus specimens. In contrast, urine primarily contained taxa that were not abundant in coronal sulcus specimens [87].
Determinants of Male Genital Microbiota Structure and Dynamics
Significant concordance in taxa found in the genital microbiota has been seen between male and female partners [90, 91]. Men whose female partners have Nugent-BV are more likely to carry BV-associated anaerobes such as Gardnerella and Prevotella, while those whose female partners do not have BV are more likely to carry Lactobacillus spp., as well as Corynebacterium and Staphylococcus [90]. Studies characterizing G. vaginalis clades detected in the genital microbiota of monogamous heterosexual couples show that they share the same strains [92]. These data suggest an important role for sexual activity in determining the male (and female) genital microbiota, provide support for the sexual transmissibility of BV and support male partner treatment clinical trials to eliminate BV recurrence [93]. A recent study following partners’ genital microbiota showed that baseline penile microbiota predicted BV incidence in women who did not have BV at baseline, supporting the critical contribution of the penile microbiota to the composition of the vaginal microbiota [94].
Anatomy is also a major determinant of the genital microbiota in men; the foreskin represents a unique physical and biochemical environment that harbors a specific microbiota different from that of the corona sulcus, and removal of the foreskin during male circumcision causes dramatic changes in the penile microbiota (Table 2). Uncircumcised men have high penile bacterial density and high absolute abundances of anaerobic bacteria, including many which are associated with BV [87, 89, 90, 95]. However, after the foreskin is removed, penile microbiota density and microbiota diversity decline, anaerobes decrease significantly, and the genital microbiota becomes dominated by members of the genera Staphylococcus and Corynebacterium, but in low absolute abundances [89, 95]. Interestingly, male circumcision has been shown to reduce HIV, herpes simplex 2 (HSV2), and human papillomavirus (HPV) acquisition in men [96–99] and decreased BV in female partners [76]. The relationship is bidirectional, as BV increases the risk of HIV acquisition by a male partner [100]. Finally, while rapid short-term fluctuations in the vaginal microbiota are well documented [43, 49], aside from the major changes associated with circumcision, little is known about temporal dynamics and stability of the penile and/or urethral microbiota.
Table 2.
Factors Associated With Male Genital Microbiota Structure
Increased Anaerobes/BV-Associated Bacteria | Decreased Anaerobes/BV-Associated Bacteria |
---|---|
Uncircumcised penis | Circumcised penis |
Female partner with BV | Female partner without BV |
Abbreviation: BV, bacterial vaginosis.
EPIDEMIOLOGIC ASSOCIATIONS BETWEEN GENITAL MICROBIOTA AND SEXUALLY TRANSMITTED INFECTIONS
In women, having Amsel-BV, Nugent-BV, or molecular-BV [26] (as compared to a Lactobacillus-dominated vaginal microbiota) has been linked to an increased risk for the acquisition of most sexually transmitted infections, including gonorrhea, chlamydia, trichomoniasis, HPV, HSV, Mycoplasma genitalium, as well as acquisition and transmission of HIV [7, 8, 31, 35, 100–104]. Longitudinal studies have shown that BV in general is associated with increased risk of incident cervicovaginal chlamydia, gonorrhea, and trichomonas infection [101, 105–108].
A strong association exists between BV (as measured by Amsel’s criteria, Nugent score, or molecularly) and prevalent and incident HIV infection [6, 7]. Specific bacteria associated with BV, including Prevotella and Sneathia amnii, have been shown to be associated with increased risk of HIV [6], as well as elevated concentrations of the BV-associated bacteria Parvimonas, Gemella asaccharolytica, Mycoplasma hominis, Leptotrichia/Sneathia, Eggerthella spp. type 1, and Megasphaera [109].
L. iners-dominated vaginal microbiota may place women at increased risk for Chlamydia trachomatis and HIV acquisition relative to those with vaginal microbiota dominated by L. crispatus [6, 110, 111]. In a prospective study in South Africa, no woman with L. crispatus-dominated vaginal microbiota acquired HIV [6].
Much less is known about epidemiologic associations between the penile microbiota and STIs. One study showed that men with asymptomatic STIs (gonorrhea and chlamydia) were more likely to have urine microbiota dominated by fastidious, anaerobic, and uncultivated bacteria (potentially reflective of urethral colonization) than those without STI [12]. Previous cultivation-based studies have shown similar results [12]. Finally, the presence of high densities of anaerobic bacteria on the penis has been associated with increased risk of HIV acquisition [13].
MECHANISMS BEHIND GENITAL MICROBIOTA-MEDIATED RISK OR PROTECTION FROM STIS
Female Genital Microbiota
The mechanisms by which the female genital microbiota may enhance or attenuate risk of STI acquisition have been much more extensively studied than those of the male genital microbiota, although many research gaps remain. Vaginal lactobacilli are thought to protect against STI pathogens through several broad mechanisms, including competitive binding to vaginal epithelial cells [112], the production of bacteriocins, direct inhibition of pathogens through metabolite production [113, 114], modulation of the pathogen-host cell interaction [115], and through impacts on the host immune system, that is through controlling inflammation in the vaginal microenvironment [116]. We briefly discuss Lactobacillus competitive binding, lactic acid production, and selected mechanisms by which BV-associated bacteria may impact STI susceptibility in more detail below.
Studies have shown that some Lactobacillus strains isolated from the vaginal tract can bind tightly to vaginal epithelial cells, and prevent Neisseria gonorrhoeae attachment and cell invasion as well as attachment of some BV-associated bacteria (eg, G. vaginalis or Prevotella bivia), perhaps through competitive binding [112, 117, 118]. Similarly, Trichomonas vaginalis, a parasite that requires adherence to the vaginal epithelium to induced cytopathic effects [119], can be prevented from binding by strains of L. jensenii and L. gasseri [120]. The effect was dependent on cell contact, although the exact mechanisms remains unclear [120]. It is important to note that this parasite grows more efficiently at high pH, so the acidic vaginal environment associated with Lactobacillus-dominated microbiota may contribute to growth inhibition [121].
Importantly, many of the protective effects seem to be mediated, at least in part, through the action of a key metabolite produced by lactobacilli, lactic acid. Lactobacillus spp. have been shown to inhibit the growth of N. gonorrhoeae due to the effects of acidification by lactic acid [122]. Lactic acid, and not low pH alone, produced by Lactobacillus spp. has been shown in vitro to inactivate C. trachomatis as well [113, 123]. Lactic acid has 2 isoforms, d(−) and l(+) lactic acid [115, 124]. L. crispatus and L. gasseri produce both d(−) and l(+) isomers of lactic acid, L. iners produces only l(+)lactic acid, and L. jensenii only d(−) lactic acid, while human cells only make l(+) lactic acid [114, 125]. Accumulating evidence suggests that d(−) lactic acid may be more protective (through a variety of mechanisms) against STIs, including chlamydia and HIV, than l(+) lactic acid [20, 115]. Interestingly, this effect appeared to be mediated by effects on the host cell. Inhibition of epithelial cell migration and proliferation by d(−) lactic acid and L. crispatus or L. jensenii culture supernatants afford a strong inhibition of C. trachomatis infection of these cells [126]. This may in part be why epidemiologic studies show an increased risk of acquisition of chlamydia and to a lesser extent HIV in women with L. iners-dominated microbiota as compared to vaginal microbiota dominated by other Lactobacillus spp. In addition, lactic acid has been shown to have anti-inflammatory properties, which likely contributes to its beneficial effect, including protection against HIV [116, 127, 128]. A full review of the potential mechanisms by which the vaginal microbiota may impact HIV acquisition is outside of the scope of this review, and we refer readers to excellent reviews by Eastment et al [126] and Petrova et al [129].
Conversely, BV-associated bacteria may create a permissive environment for STI pathogen invasion mediated by elevated pH, enhanced inflammation, and through the production of metabolites, that is indole and biogenic amines, which may facilitate STI infection [26, 130–132]. For example, upon infection C. trachomatis stimulates an interferon-γ cascade in the host cell, which, through a series of enzymatic reactions, converts tryptophan to kynurenine, depleting intracellular pools of tryptophan. As Chlamydia is auxotrophic for tryptophan, this is an important defense against the pathogen. Interestingly, some strains of BV-associated bacteria (eg, Prevotella) are capable of producing indole, which C. trachomatis is able to convert to tryptophan, resulting in “rescue” of the pathogen [133]. Lastly, biogenic amines, which are elevated in women with BV, are associated with odor, but more importantly, have been shown to increase the virulence of N. gonorrhoeae, protecting the bacterium from innate immune defenses [131, 134, 135].
Male Genital Microbiota
Little is known regarding potential mechanisms relating male genital microbiota to STI susceptibility. However, there is a small amount of preliminary data relating to HIV. An analysis of samples collected during a male circumcision trial conducted in Rakai, Uganda, demonstrated that amongst 182 uncircumcised men, the 46 men who became HIV infected during the course of the trial had significantly higher abundances of penile anaerobes at study baseline (including Prevotella, Dialister, Mobiluncus, Murdochiella, and Peptostreptococcus) than those who did not become infected. These anaerobic bacteria were amongst those that had previously been found to be decreased after circumcision. Increased abundances of anaerobic bacteria correlated with elevated proinflammatory cytokines from coronal sulcus swabs, most notably interleukin-8 (IL-8) [13]. Thus, it is hypothesized that colonization with BV-associated organisms may lead to increased inflammation in the male genital tract, hence leading to increased susceptibility to HIV.
KNOWLEDGE GAPS AND NEED FOR NEW STUDIES
While the relationship between the male and female genital microbiota and STI risk is an area of active research, many knowledge gaps remain. The association between the male genital microbiota and HIV risk has only begun to be studied, and relationships to other STIs are largely unknown. Furthermore, whether the microbiota of the coronal sulcus (on which most existing studies are based) or that of the urethra plays a greater role in STI vulnerability is unclear.
Many early epidemiologic studies linking STIs and the vaginal microbiota have been cross-sectional, making causal inferences difficult. Increasingly, longitudinal studies are assessing the vaginal microbiota prior to infection with STI acquisition [6, 101], but there is still a need for more research in this area. Moreover, given the potential for significant short-term fluctuation in the vaginal microbiota and the difficulty of ascertaining exactly when an infection occurs, considerable uncertainty remains in trying to assess the relationship between the vaginal microbiota based on a single vaginal swab (even one collected prior to infection) to STI risk. It is possible that vaginal microbiota instability from Lactobacillus-dominated to high abundance of anaerobes is an important risk factor for STI acquisition. The frequency and duration of these periods of paucity in Lactobacillus spp. might represent better the actual risk. While more research to understand factors associated with stability of the vaginal microbiota and STI risk is needed, study design is difficult given that frequent longitudinal sampling prior to infection would be required.
As molecular techniques are becoming more widespread and affordable, comparison to earlier methods of assessing the vaginal microbiota would help assess their benefit as predictors of STI risk. For example, older studies relying on the Amsel criteria or Nugent score to diagnose BV have estimated effect sizes (ie, ≤2-fold) [7] for HIV risk that are smaller than those measured in a more recent study (Gosmann et al) utilizing new molecular techniques (ie, >4-fold) [6, 7, 26, 109]. This may indicate that molecular-BV, which more broadly characterizes the vaginal microbial community, may be more appropriate to use in studies to understand the potential causal relationship between cervicovaginal microbiota and HIV acquisition. However, further studies are required to better understand why these differences are observed and how these differences may help to develop improved STI prevention approaches. Current US guidelines recommend treatment only for women with symptomatic BV (representing about 50% of those with BV [30]), and syndromic management of STI risk in many low-resource settings, including much of Africa, is common. However, the relative risk for STIs or HIV in women with symptomatic versus asymptomatic BV is unknown. Existing studies examining the relationship between BV and STIs or HIV have used a composite BV measurement, which includes both symptomatic and asymptomatic women [7, 31].
Knowledge gaps remain in our understanding of the differential contribution of the overall structure of the vaginal microbiota [111] versus key species [108, 109] in STI risk. Moreover, the relative contribution of absolute bacterial load (the quantities of bacteria) in the vaginal microbiota (both overall and of key bacterial species), relative abundance of bacteria, and the potential contribution of low-abundance species to STI risk will require additional research. While new technologies such as 16S rRNA sequencing techniques have provided important information on individual species present in the vaginal microbiota, even this level of detail may be insufficient. Information on specific strains of bacteria and their metabolic products and immunologic interactions may be necessary, as individual strains or combination of strains [136] of the same species of bacteria may lead to different phenotypes with differing implications for health and disease. Relatively little is known about the interactions between bacterial members of the vaginal microbiota [137, 138], and whether the mycobiome [139, 140] and virome [139, 140] of the vaginal microenvironment may relate to STI risk and protection. Finally, the vaginal microenvironment is a complex result of interactions between the endogenous microbiota, host immunity, exogenous and endogenous hormones, and exposures (related to sex, menses, or hygiene practices). Studies evaluating all aspects of the vaginal microenvironment and their relative contribution to STI vulnerability are desperately needed.
CONSIDERATIONS FOR DEVELOPING STI PREVENTIVE STRATEGIES
An important long-term goal of research on the genital microbiota and STI protection or risk is to develop novel strategies to manipulate and optimize the microbiota to prevent STI acquisition and transmission. In terms of the male genital microbiota, circumcision has been extensively studied and has already been implemented as an intervention to reduce HIV risk. Studies as outlined above show that at least some of this protection may result from decreases in anaerobes in the penile microbiota after circumcision [13]. These findings have led to the development of a randomized open-label trial to treat uncircumcised men who plan to undergo circumcision with oral tinidazole, penile topical metronidazole, topical clindamycin, or topical hydrogen peroxide for a month prior to circumcision in order to assess whether these may decrease HIV entry into foreskin-derived CD4 T cells at the time of circumcision [141]. If promising, these studies could lead to new topical interventions to prevent HIV in men who are not interested in undergoing circumcision.
To date, options are limited to manipulate the penile microbiota, and our understanding of its function in sexual health is nascent. However, several strategies are being considered to modulate the composition and function of the vaginal microbiota. Development and implementation of these approaches involves multiple considerations. In the case of a drug, it is critical to understand how its delivery to the vagina would affect the composition of the vaginal microbiota and, more importantly, if metabolism of the resident microbiota may affect the efficacy of the medications to prevent [142] or treat [143] STIs. Further, while low-Lactobacillus microbiota, and to a lesser extent L. iners-dominated vaginal microbiota, have been associated with STI acquisition and poor obstetric outcomes, whether the optimal microbiota is the same for each woman is not necessarily clear. Only 50% of women with low-Lactobacillus vaginal microbiota experience symptoms, while STI acquisition is ultimately a function not only of risk but of exposure. In the absence of substantial STI exposure, symptoms, or pregnancy, it is unclear to what degree having a low-Lactobacillus vaginal microbiota represents a “pathogenic” state, which should be altered, or whether there may be some as-yet unappreciated benefits, for example to women of African descent, in whom asymptomatic BV is much more prevalent. Thus, modulating the microbiota should be considered with caution until we improve our understanding of the contribution of all vaginal microenvironment types in health and disease.
One of the greatest challenges associated with modulation of the vaginal microbiota to prevent STIs is the difficulty in establishing lasting alterations to the vaginal microbiota composition and function. In women with symptomatic BV, to date, effecting a lasting cure of symptoms and “reset” of the vaginal microbiota to a Lactobacillus-dominated state has been elusive. While after antibiotic therapy vaginal lactobacilli initially increase and anaerobes decrease, over 50% of women with symptomatic BV experience recurrence with anaerobic overgrowth within 6 months, and a subset recur repeatedly, leading to significant patient frustration, lower quality of life, and morbidity [144, 145]. In one preliminary study of periodic presumptive therapy conducted in the United States and Kenya, women receiving monthly high-dose intravaginal metronidazole suppositories had a lower incidence of a composite measurement of any bacterial STIs (C. trachomatis, N. gonorrhoeae, or M. genitalium). However, when assessed individually, reductions in STI incidences were not statistically significant [146], and the reduction in the proportion of visits with BV in the treatment arm when compared to placebo (21% vs 33%) was relatively modest, although statistically significant [147]. Another trial showed decreases in incident chlamydia in women with asymptomatic BV randomized to twice-weekly metronidazole vaginal gel compared to women receiving standard of care [148]. However, a third trial of home screening and treatment of women with asymptomatic BV (screening was conducted every 2 months for 12 months, and women with BV were randomized to treatment with oral metronidazole 500 mg twice daily for 7 days or observation alone) did not result in a reduction in incidence of gonorrhea or chlamydia [149]. In sum, results of periodic presumptive therapy or suppressive therapy to prevent STIs are mixed. In addition, there are potential side effects, including the development of antibiotic resistance to suppressive regimens. Reductions in BV even while on therapy may be relatively modest and, critically, there is no evidence of a sustained effect of these regimens as after cessation of therapy, BV typically quickly recurs [150]. The lack of efficacious treatment options hampers our ability to modulate risk of STIs.
Moreover, while treating BV does seem to reduce, at least temporarily, the levels of some inflammatory cytokines associated with HIV acquisition, such as IL-1β, the picture may be more complex. A recent study showed that while BV treatment reduced IL-1β, as anaerobes decreased and L. iners increased, higher levels of several other genital chemokines associated with HIV acquisition, including IP-10, MIP-3a, and MIG, were observed [151].
A highly promising option for vaginal microbiota manipulation is through the application of live biotherapeutic products aimed at restoring Lactobacillus-dominated microbiota. Trials of such products have been conducted either alone or together with antibiotics or other adjunctive therapies to treat BV. Many of these have been small, the tested products were diverse, and results have been mixed with insufficient evidence to recommend routine use of any one product [130]. However, a recent randomized placebo-controlled trial compared periodic vaginal repletion with a human L. crispatus strain (LACTIN-V), and demonstrated a modest reduction in recurrent BV among those who used the active study product [152]—a promising finding that should reenergize this field and lead to the development of novel and rationally designed live biotherapeutics products that consist of a community of bacteria, similarly to innovations in manipulating the gut microbiota [153]. Finally, new therapies are being pioneered: a recent small case series reported 5 women who received vaginal microbiota transplants for recurrent BV [154]. This promising experimental therapy with unique ethical issues and that potentially carries risks, is expected to yield critical information to improve and design novel live biotherapeutic products. In addition, the use of novel agents to disrupt the tenacious biofilm that characterizes BV might offer another avenue for intervention: in a small study, the boric acid and EDTA-based intravaginal product TOL-463 had some efficacy for treatment of for both acute BV and vulvovaginal candidiasis, and is currently under study for the prevention of recurrent BV [155] (Clinical Trial Registration NCT03930745).
In the face of rising STI rates and challenges surrounding antibiotic resistance, antibiotic-sparing approaches to prevent STIs are needed. For the long term, novel strategies to improve reproductive health in women, and potentially in men, around the world are being developed. These innovative therapies might involve combinations of more promising interventions, including transplants of healthy vaginal fluid, synthetic or rationally assembled consortia of bacteria (next-generation live biotherapeutics), small molecules that target biofilm or specific pathogens, and the combined treatment of partners targeting both the male and female microbiota at the same time.
Notes
Financial support. This work was supported by the National Institute of Allergy and Infectious Diseases and the National Institute of Nursing Research, National Institutes of Health (grant numbers U19AI08044 and R01NR015495 to J. R. and K23AI125715 to S. T.).
Supplement sponsorship. This work is part of a supplement sponsored by the National Institute of Allergy and Infectious Diseases (NIAID), part of the National Institutes of Health (NIH) and the Centers for Disease Control and Prevention (CDC).
Potential conflicts of interest. J. R. is a cofounder of LUCA Biologics, a biotechnology company focusing on translating microbiome research into live biotherapeutic drugs for women’s health. S. T. has served as a consultant for LUCA Biologics, Biofire Diagnostics, and Roche Molecular Diagnostics, and has received speaker honoraria from Roche Molecular Diagnostics. J. M. is a member of the Scientific Advisory Board for OSEL, the manufacturer of LACTIN-V, and has received research support and served as a scientific advisor for vaginal health research from BD. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
References
- 1. Centers for Disease Control and Prevention. Sexually transmitted disease surveillance 2018. Atlanta, GA: US Department of Health and Human Services, 2019. [Google Scholar]
- 2. World Health Organization (WHO). Report on global sexually transmitted infection surveillance 2018. Geneva, Switzerland: WHO, 2018. [Google Scholar]
- 3. Unemo M, Jensen JS. Antimicrobial-resistant sexually transmitted infections: gonorrhoea and Mycoplasma genitalium. Nat Rev Urol 2017; 14:139–52. [DOI] [PubMed] [Google Scholar]
- 4. Petrova MI, Lievens E, Malik S, Imholz N, Lebeer S. Lactobacillus species as biomarkers and agents that can promote various aspects of vaginal health. Front Physiol 2015; 6:81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Martin DH. The microbiota of the vagina and its influence on women’s health and disease. Am J Med Sci 2012; 343:2–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. 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; 46:29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Atashili J, Poole C, Ndumbe PM, Adimora AA, Smith JS. Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. AIDS 2008; 22:1493–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Brotman RM. Vaginal microbiome and sexually transmitted infections: an epidemiologic perspective. J Clin Invest 2011; 121:4610–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Elovitz MA, Gajer P, Riis V, et al. Cervicovaginal microbiota and local immune response modulate the risk of spontaneous preterm delivery. Nat Commun 2019; 10:1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Fettweis JM, Serrano MG, Brooks JP, et al. The vaginal microbiome and preterm birth. Nat Med 2019; 25:1012–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hillier SL, Nugent RP, Eschenbach DA, et al. Association between bacterial vaginosis and preterm delivery of a low-birth-weight infant. The Vaginal Infections and Prematurity Study Group. N Engl J Med 1995; 333:1737–42. [DOI] [PubMed] [Google Scholar]
- 12. Nelson DE, Van Der Pol B, Dong Q, et al. Characteristic male urine microbiomes associate with asymptomatic sexually transmitted infection. PLoS One 2010; 5:e14116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Liu CM, Prodger JL, Tobian AAR, et al. Penile anaerobic dysbiosis as a risk factor for HIV infection. mBio 2017; 8:e00996-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Prodger JL, Kaul R. The biology of how circumcision reduces HIV susceptibility: broader implications for the prevention field. AIDS Res Ther 2017; 14:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Holmes KK, Sparling FP, Stamm WE, et al. Host Immunity and molecular pathogenesis and STD. In: Sexually transmitted diseases. 4th ed. New York: McGraw-Hill Medical, 2007:269–320. [Google Scholar]
- 16. Brotman RM, Ravel J, Bavoil PM, Gravitt PE, Ghanem KG. Microbiome, sex hormones, and immune responses in the reproductive tract: challenges for vaccine development against sexually transmitted infections. Vaccine 2014; 32:1543–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Yi TJ, Shannon B, Prodger J, McKinnon L, Kaul R. Genital immunology and HIV susceptibility in young women. Am J Reprod Immunol 2013; 69 (Suppl 1):74–9. [DOI] [PubMed] [Google Scholar]
- 18. Vitali D, Wessels JM, Kaushic C. Role of sex hormones and the vaginal microbiome in susceptibility and mucosal immunity to HIV-1 in the female genital tract. AIDS Res Ther 2017; 14:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Schroeder HA, Nunn KL, Schaefer A, et al. Herpes simplex virus-binding IgG traps HSV in human cervicovaginal mucus across the menstrual cycle and diverse vaginal microbial composition. Mucosal Immunol 2018; 11:1477–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Nunn KL, Wang YY, Harit D, et al. Enhanced trapping of HIV-1 by human cervicovaginal mucus is associated with Lactobacillus crispatus-dominant microbiota. mBio 2015; 6:e01084-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Wessels JM, Felker AM, Dupont HA, Kaushic C. The relationship between sex hormones, the vaginal microbiome and immunity in HIV-1 susceptibility in women. Dis Model Mech 2018; 11:dmm035147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Ceccarani C, Marangoni A, Severgnini M, et al. Rectal microbiota associated with Chlamydia trachomatis and Neisseria gonorrhoeae infections in men having sex with other men. Front Cell Infect Microbiol 2019; 9:358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Döderlein A. Das Scheidensekret und seine Bedeutung für das Puerperalfieber, 1892. Norderstedt, Germany: Books on Demand, 2012. [reproduction]. [Google Scholar]
- 24. Witkin SS, Linhares IM. Why do lactobacilli dominate the human vaginal microbiota? BJOG 2017; 124:606–11. [DOI] [PubMed] [Google Scholar]
- 25. Martin DH, Marrazzo JM. The vaginal microbiome: current understanding and future directions. J Infect Dis 2016; 214 (Suppl 1):S36–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. McKinnon LR, Achilles SL, Bradshaw CS, et al. The evolving facets of bacterial vaginosis: implications for HIV transmission. AIDS Res Hum Retroviruses 2019; 35:219–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kenyon C, Colebunders R, Crucitti T. The global epidemiology of bacterial vaginosis: a systematic review. Am J Obstet Gynecol 2013; 209:505–23. [DOI] [PubMed] [Google Scholar]
- 28. Peebles K, Velloza J, Balkus JE, McClelland RS, Barnabas RV. High global burden and costs of bacterial vaginosis: a systematic review and meta-analysis. Sex Transm Dis 2019; 46:304–11. [DOI] [PubMed] [Google Scholar]
- 29. Amsel R, Totten PA, Spiegel CA, Chen KC, Eschenbach D, Holmes KK. Nonspecific vaginitis. Diagnostic criteria and microbial and epidemiologic associations. Am J Med 1983; 74:14–22. [DOI] [PubMed] [Google Scholar]
- 30. Workowski KA. Centers for disease control and prevention sexually transmitted diseases treatment guidelines. Clin Infect Dis 2015; 61 (Suppl 8):S759–62. [DOI] [PubMed] [Google Scholar]
- 31. Tamarelle J, Thiébaut ACM, de Barbeyrac B, Bébéar C, Ravel J, Delarocque-Astagneau E. The vaginal microbiota and its association with human papillomavirus, Chlamydia trachomatis, Neisseria gonorrhoeae and Mycoplasma genitalium infections: a systematic review and meta-analysis. Clin Microbiol Infect 2019; 25:35–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Sha BE, Chen HY, Wang QJ, Zariffard MR, Cohen MH, Spear GT. Utility of Amsel criteria, Nugent score, and quantitative PCR for Gardnerella vaginalis, Mycoplasma hominis, and Lactobacillus spp. for diagnosis of bacterial vaginosis in human immunodeficiency virus-infected women. J Clin Microbiol 2005; 43:4607–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Coleman JS, Gaydos CA. Molecular diagnosis of bacterial vaginosis: an update. J Clin Microbiol 2018; 56:e00342-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Achilles SL, Austin MN, Meyn LA, Mhlanga F, Chirenje ZM, Hillier SL. Impact of contraceptive initiation on vaginal microbiota. Am J Obstet Gynecol 2018; 218:622.e1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Brotman RM, Bradford LL, Conrad M, et al. Association between Trichomonas vaginalis and vaginal bacterial community composition among reproductive-age women. Sex Transm Dis 2012; 39:807–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Brotman RM, He X, Gajer P, et al. Association between cigarette smoking and the vaginal microbiota: a pilot study. BMC Infect Dis 2014; 14:471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Brotman RM, Shardell MD, Gajer P, et al. Association between the vaginal microbiota, menopause status, and signs of vulvovaginal atrophy. Menopause 2014; 21:450–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Brotman RM, Shardell MD, Gajer P, et al. Interplay between the temporal dynamics of the vaginal microbiota and human papillomavirus detection. J Infect Dis 2014; 210:1723–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Brown RG, Marchesi JR, Lee YS, et al. Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med 2018; 16:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Callahan BJ, DiGiulio DB, Goltsman DSA, et al. Replication and refinement of a vaginal microbial signature of preterm birth in two racially distinct cohorts of US women. Proc Natl Acad Sci U S A 2017; 114:9966–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Chaban B, Links MG, Jayaprakash TP, et al. Characterization of the vaginal microbiota of healthy Canadian women through the menstrual cycle. Microbiome 2014; 2:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Datcu R, Gesink D, Mulvad G, et al. Vaginal microbiome in women from Greenland assessed by microscopy and quantitative PCR. BMC Infect Dis 2013; 13:480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Gajer P, Brotman RM, Bai G, et al. Temporal dynamics of the human vaginal microbiota. Sci Transl Med 2012; 4:132ra52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Ravel J, Gajer P, Abdo Z, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A 2011; 108 (Suppl 1):4680–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Reimers LL, Mehta SD, Massad LS, et al. The cervicovaginal microbiota and its associations with human papillomavirus detection in HIV-infected and HIV-uninfected women. J Infect Dis 2016; 214:1361–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Romero R, Hassan SS, Gajer P, et al. The vaginal microbiota of pregnant women who subsequently have spontaneous preterm labor and delivery and those with a normal delivery at term. Microbiome 2014; 2:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Van Der Pol B, Williams JA, Fuller D, Taylor SN, Hook EW 3rd. Combined testing for Chlamydia, Gonorrhea, and Trichomonas by use of the BD max CT/GC/TV assay with genitourinary specimen types. J Clin Microbiol 2017; 55:155–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Van Der Pol B, Daniel G, Kodsi S, Paradis S, Cooper CK. Molecular-based testing for sexually transmitted infections using samples previously collected for vaginitis diagnosis. Clin Infect Dis 2019; 68:375–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Srinivasan S, Liu C, Mitchell CM, et al. Temporal variability of human vaginal bacteria and relationship with bacterial vaginosis. PLoS One 2010; 5:e10197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. France MT, Ma B, Gajer P, et al. VALENCIA: a nearest centroid classification method for vaginal microbial communities based on composition. Microbiome 2020; 8:166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Spear GT, French AL, Gilbert D, et al. Human α-amylase present in lower-genital-tract mucosal fluid processes glycogen to support vaginal colonization by Lactobacillus. J Infect Dis 2014; 210:1019–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. van der Veer C, Hertzberger RY, Bruisten SM, et al. Comparative genomics of human Lactobacillus crispatus isolates reveals genes for glycosylation and glycogen degradation: implications for in vivo dominance of the vaginal microbiota. Microbiome 2019; 7:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Farage M, Miller KW, Sobel J. Dynamics of the vaginal ecosystem—hormonal influences . Infect Dis 2010; 3:doi: 10.4137/IDRT.S3903. [DOI] [Google Scholar]
- 54. Widholm O, Kantero RL, Axelson E, Johansson ED, Wide L. Endocrine changes before and after the menarche. I. Urinary excretion of estrogen, FSH and LH, and serum levels of progesterone, FSH and LH. Acta Obstet Gynecol Scand 1974; 53:197–208. [DOI] [PubMed] [Google Scholar]
- 55. Thoma ME, Gray RH, Kiwanuka N, et al. Longitudinal changes in vaginal microbiota composition assessed by gram stain among never sexually active pre- and postmenarcheal adolescents in Rakai, Uganda. J Pediatr Adolesc Gynecol 2011; 24:42–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Hickey RJ, Zhou X, Settles ML, et al. Vaginal microbiota of adolescent girls prior to the onset of menarche resemble those of reproductive-age women. mBio 2015; 6:e00097-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Mirmonsef P, Hotton AL, Gilbert D, et al. Free glycogen in vaginal fluids is associated with Lactobacillus colonization and low vaginal pH. PLoS One 2014; 9:e102467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Mirmonsef P, Modur S, Burgad D, et al. Exploratory comparison of vaginal glycogen and Lactobacillus levels in premenopausal and postmenopausal women. Menopause 2015; 22:702–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Eschenbach DA, Thwin SS, Patton DL, et al. Influence of the normal menstrual cycle on vaginal tissue, discharge, and microflora. Clin Infect Dis 2000; 30:901–7. [DOI] [PubMed] [Google Scholar]
- 60. Mitchell CM, Srinivasan S, Zhan X, et al. Vaginal microbiota and genitourinary menopausal symptoms: a cross-sectional analysis. Menopause 2017; 24:1160–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Nunn KL, Ridenhour BJ, Chester EM, Vitzthum VJ, Fortenberry JD, Forney LJ. Vaginal glycogen, not estradiol, is associated with vaginal bacterial community composition in black adolescent women. J Adolesc Health 2019; 65:130–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Freitas AC, Chaban B, Bocking A, et al. ; VOGUE Research Group . The vaginal microbiome of pregnant women is less rich and diverse, with lower prevalence of mollicutes, compared to non-pregnant women. Sci Rep 2017; 7:9212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Walther-António MR, Jeraldo P, Berg Miller ME, et al. Pregnancy’s stronghold on the vaginal microbiome. PLoS One 2014; 9:e98514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Shen J, Song N, Williams CJ, et al. Effects of low dose estrogen therapy on the vaginal microbiomes of women with atrophic vaginitis. Sci Rep 2016; 6:24380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Crucitti T, Hardy L, van de Wijgert J, et al. ; Ring Plus Study Group . 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] [PMC free article] [PubMed] [Google Scholar]
- 66. Kriszt R, Winkler Z, Polyák Á, et al. Xenoestrogens ethinyl estradiol and zearalenone cause precocious puberty in female rats via central kisspeptin signaling. Endocrinology 2015; 156:3996–4007. [DOI] [PubMed] [Google Scholar]
- 67. .Fettweis JM, Brooks JP, Serrano MG, et al. Differences in vaginal microbiome in African American women versus women of European ancestry. Microbiology (Reading) 2014; 160:2272–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Zhou X, Brown CJ, Abdo Z, et al. Differences in the composition of vaginal microbial communities found in healthy Caucasian and black women. ISME J 2007; 1:121–33. [DOI] [PubMed] [Google Scholar]
- 69. Allsworth JE, Peipert JF. Prevalence of bacterial vaginosis: 2001–2004 National Health and Nutrition Examination Survey data. Obstet Gynecol 2007; 109:114–20. [DOI] [PubMed] [Google Scholar]
- 70. Paul K, Boutain D, Manhart L, Hitti J. Racial disparity in bacterial vaginosis: the role of socioeconomic status, psychosocial stress, and neighborhood characteristics, and possible implications for preterm birth. Soc Sci Med 2008; 67:824–33. [DOI] [PubMed] [Google Scholar]
- 71. Klebanoff MA, Andrews WW, Zhang J, et al. Race of male sex partners and occurrence of bacterial vaginosis. Sex Transm Dis 2010; 37:184–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Brookheart RT, Lewis WG, Peipert JF, Lewis AL, Allsworth JE. Association between obesity and bacterial vaginosis as assessed by Nugent score. Am J Obstet Gynecol 2019; 220:476.e1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Tuddenham S, Ghanem KG, Caulfield LE, et al. Associations between dietary micronutrient intake and molecular-bacterial vaginosis. Reprod Health 2019; 16:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lokken EM, Richardson BA, Kinuthia J, et al. A prospective cohort study of the association between body mass index and incident bacterial vaginosis. Sex Transm Dis 2019; 46:31–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Koumans EH, Sternberg M, Bruce C, et al. The prevalence of bacterial vaginosis in the United States, 2001–2004; associations with symptoms, sexual behaviors, and reproductive health. Sex Transm Dis 2007; 34:864–9. [DOI] [PubMed] [Google Scholar]
- 76. Gray RH, Kigozi G, Serwadda D, et al. The effects of male circumcision on female partners’ genital tract symptoms and vaginal infections in a randomized trial in Rakai, Uganda. Am J Obstet Gynecol 2009; 200:42.e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Borovkova N, Korrovits P, Ausmees K, et al. Influence of sexual intercourse on genital tract microbiota in infertile couples. Anaerobe 2011; 17:414–8. [DOI] [PubMed] [Google Scholar]
- 78. Brotman RM, Ravel J, Cone RA, Zenilman JM. Rapid fluctuation of the vaginal microbiota measured by Gram stain analysis. Sex Transm Infect 2010; 86:297–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Brotman RM, Klebanoff MA, Nansel TR, et al. A longitudinal study of vaginal douching and bacterial vaginosis–a marginal structural modeling analysis. Am J Epidemiol 2008; 168:188–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Marrazzo JM, Koutsky LA, Eschenbach DA, Agnew K, Stine K, Hillier SL. Characterization of vaginal flora and bacterial vaginosis in women who have sex with women. J Infect Dis 2002; 185:1307–13. [DOI] [PubMed] [Google Scholar]
- 81. Forcey DS, Vodstrcil LA, Hocking JS, et al. Factors associated with bacterial vaginosis among women who have sex with women: a systematic review. PLoS One 2015; 10:e0141905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Fethers K, Twin J, Fairley CK, et al. Bacterial vaginosis (BV) candidate bacteria: associations with BV and behavioural practices in sexually-experienced and inexperienced women. PLoS One 2012; 7:e30633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Marrazzo JM, Thomas KK, Fiedler TL, Ringwood K, Fredricks DN. Risks for acquisition of bacterial vaginosis among women who report sex with women: a cohort study. PLoS One 2010; 5:e11139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Marrazzo JM, Thomas KK, Agnew K, Ringwood K. Prevalence and risks for bacterial vaginosis in women who have sex with women. Sex Transm Dis 2010; 37:335–9. [PMC free article] [PubMed] [Google Scholar]
- 85. Tamarelle J, Ma B, Gajer P, et al. Nonoptimal vaginal microbiota after azithromycin treatment for Chlamydia trachomatis infection. J Infect Dis 2020; 221:627–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Ravel J, Brotman RM, Gajer P, et al. Daily temporal dynamics of vaginal microbiota before, during and after episodes of bacterial vaginosis. Microbiome 2013; 1:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Nelson DE, Dong Q, Van der Pol B, et al. Bacterial communities of the coronal sulcus and distal urethra of adolescent males. PLoS One 2012; 7:e36298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Dong Q, Nelson DE, Toh E, et al. The microbial communities in male first catch urine are highly similar to those in paired urethral swab specimens. PLoS One 2011; 6:e19709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Price LB, Liu CM, Johnson KE, et al. The effects of circumcision on the penis microbiome. PLoS One 2010; 5:e8422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Liu CM, Hungate BA, Tobian AA, et al. Penile microbiota and female partner bacterial vaginosis in Rakai, Uganda. mBio 2015; 6:e00589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Zozaya M, Ferris MJ, Siren JD, et al. Bacterial communities in penile skin, male urethra, and vaginas of heterosexual couples with and without bacterial vaginosis. Microbiome 2016; 4:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Eren AM, Zozaya M, Taylor CM, Dowd SE, Martin DH, Ferris MJ. Exploring the diversity of Gardnerella vaginalis in the genitourinary tract microbiota of monogamous couples through subtle nucleotide variation. PLoS One 2011; 6:e26732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Muzny CA, Pontius A, Woznicki N, et al. Use of a novel couples’ verification tool in a male partner treatment study of women with recurrent bacterial vaginosis. Sex Transm Dis 2020; 47:e58–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Mehta SD, Zhao D, Green SJ, et al. The microbiome composition of a man’s penis predicts incident bacterial vaginosis in his female sex partner with high accuracy. Front Cell Infect Microbiol 2020; 10:433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Liu CM, Hungate BA, Tobian AA, et al. Male circumcision significantly reduces prevalence and load of genital anaerobic bacteria. mBio 2013; 4:e00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Tobian AA, Serwadda D, Quinn TC, et al. Male circumcision for the prevention of HSV-2 and HPV infections and syphilis. N Engl J Med 2009; 360:1298–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Auvert B, Taljaard D, Lagarde E, Sobngwi-Tambekou J, Sitta R, Puren A. Randomized, controlled intervention trial of male circumcision for reduction of HIV infection risk: the ANRS 1265 trial. PLoS Med 2005; 2:e298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Gray RH, Kigozi G, Serwadda D, et al. Male circumcision for HIV prevention in men in Rakai, Uganda: a randomised trial. Lancet 2007; 369:657–66. [DOI] [PubMed] [Google Scholar]
- 99. Bailey RC, Moses S, Parker CB, et al. Male circumcision for HIV prevention in young men in Kisumu, Kenya: a randomised controlled trial. Lancet 2007; 369:643–56. [DOI] [PubMed] [Google Scholar]
- 100. 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 Med 2012; 9:e1001251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Balkus JE, Richardson BA, Rabe LK, et al. Bacterial vaginosis and the risk of Trichomonas vaginalis acquisition among HIV-1-negative women. Sex Transm Dis 2014; 41:123–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Cherpes TL, Meyn LA, Krohn MA, Lurie JG, Hillier SL. Association between acquisition of herpes simplex virus type 2 in women and bacterial vaginosis. Clin Infect Dis 2003; 37:319–25. [DOI] [PubMed] [Google Scholar]
- 103. Gillet E, Meys JF, Verstraelen H, et al. Bacterial vaginosis is associated with uterine cervical human papillomavirus infection: a meta-analysis. BMC Infect Dis 2011; 11:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Lokken EM, Balkus JE, Kiarie J, et al. Association of recent bacterial vaginosis with acquisition of Mycoplasma genitalium. Am J Epidemiol 2017; 186:194–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Brotman RM, Klebanoff MA, Nansel TR, et al. Bacterial vaginosis assessed by Gram stain and diminished colonization resistance to incident gonococcal, chlamydial, and trichomonal genital infection. J Infect Dis 2010; 202:1907–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Bautista CT, Wurapa EK, Sateren WB, Hollingsworth BP, Sanchez JL. Longitudinal association of gonorrhea and bacterial vaginosis with repeat Chlamydia diagnoses among U.S. Army women: a retrospective cohort analysis. Mil Med Res 2018; 5:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Martin HL, Richardson BA, Nyange PM, et al. Vaginal lactobacilli, microbial flora, and risk of human immunodeficiency virus type 1 and sexually transmitted disease acquisition. J Infect Dis 1999; 180:1863–8. [DOI] [PubMed] [Google Scholar]
- 108. Jarrett OD, Srinivasan S, Richardson BA, et al. Specific vaginal bacteria are associated with an increased risk of Trichomonas vaginalis acquisition in women. J Infect Dis 2019; 220:1503–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. 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; 18:554–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. van der Veer C, Bruisten SM, van der Helm JJ, de Vries HJ, van Houdt R. The cervicovaginal microbiota in women notified for chlamydia trachomatis infection: a case-control study at the sexually transmitted infection outpatient clinic in Amsterdam, The Netherlands. Clin Infect Dis 2017; 64:24–31. [DOI] [PubMed] [Google Scholar]
- 111. Tamarelle J, de Barbeyrac B, Le Hen I, et al. Vaginal microbiota composition and association with prevalent Chlamydia trachomatis infection: a cross-sectional study of young women attending a STI clinic in France. Sex Transm Infect 2018; 94:616–8. [DOI] [PubMed] [Google Scholar]
- 112. Vielfort K, Sjölinder H, Roos S, Jonsson H, Aro H. Adherence of clinically isolated lactobacilli to human cervical cells in competition with Neisseria gonorrhoeae. Microbes Infect 2008; 10:1325–34. [DOI] [PubMed] [Google Scholar]
- 113. Gong Z, Luna Y, Yu P, Fan H. Lactobacilli inactivate Chlamydia trachomatis through lactic acid but not H2O2. PLoS One 2014; 9:e107758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Tachedjian G, Aldunate M, Bradshaw CS, Cone RA. The role of lactic acid production by probiotic Lactobacillus species in vaginal health. Res Microbiol 2017; 168:782–92. [DOI] [PubMed] [Google Scholar]
- 115. Edwards VL, Smith SB, McComb EJ, et al. The cervicovaginal microbiota-host interaction modulates Chlamydia trachomatis infection. mBio 2019; 10:e01548-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Aldunate M, Srbinovski D, Hearps AC, et al. Antimicrobial and immune modulatory effects of lactic acid and short chain fatty acids produced by vaginal microbiota associated with eubiosis and bacterial vaginosis. Front Physiol 2015; 6:164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Spurbeck RR, Arvidson CG. Lactobacillus jensenii surface-associated proteins inhibit Neisseria gonorrhoeae adherence to epithelial cells. Infect Immun 2010; 78:3103–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Spurbeck RR, Arvidson CG. Lactobacilli at the front line of defense against vaginally acquired infections. Future Microbiol 2011; 6:567–82. [DOI] [PubMed] [Google Scholar]
- 119. Hobbs MM, Sena AC, Swygard H, Schwebke JR. Trichomonas vaginalis and trichomoniasis. In: Holmes KK, Sparling PF, Stamm WE, et al., eds. Sexually transmitted diseases. New York: McGraw-Hill Medical, 2007:771–93. [Google Scholar]
- 120. Phukan N, Parsamand T, Brooks AE, Nguyen TN, Simoes-Barbosa A. The adherence of Trichomonas vaginalis to host ectocervical cells is influenced by lactobacilli. Sex Transm Infect 2013; 89:455–9. [DOI] [PubMed] [Google Scholar]
- 121. Petrin D, Delgaty K, Bhatt R, Garber G. Clinical and microbiological aspects of Trichomonas vaginalis. Clin Microbiol Rev 1998; 11:300–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Graver MA, Wade JJ. The role of acidification in the inhibition of Neisseria gonorrhoeae by vaginal lactobacilli during anaerobic growth. Ann Clin Microbiol Antimicrob 2011; 10:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Nardini P, Ñahui Palomino RA, Parolin C, et al. Lactobacillus crispatus inhibits the infectivity of Chlamydia trachomatis elementary bodies, in vitro study. Sci Rep 2016; 6:29024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Witkin SS, Mendes-Soares H, Linhares IM, Jayaram A, Ledger WJ, Forney LJ. Influence of vaginal bacteria and d- and l-lactic acid isomers on vaginal extracellular matrix metalloproteinase inducer: implications for protection against upper genital tract infections. mBio 2013; 4:e00460-13. [Google Scholar]
- 125. Boskey ER, Cone RA, Whaley KJ, Moench TR. Origins of vaginal acidity: high d/l lactate ratio is consistent with bacteria being the primary source. Hum Reprod 2001; 16:1809–13. [DOI] [PubMed] [Google Scholar]
- 126. Eastment MC, McClelland RS. Vaginal microbiota and susceptibility to HIV. AIDS 2018; 32:687–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Hearps AC, Tyssen D, Srbinovski D, et al. Vaginal lactic acid elicits an anti-inflammatory response from human cervicovaginal epithelial cells and inhibits production of pro-inflammatory mediators associated with HIV acquisition. Mucosal Immunol 2017; 10:1480–90. [DOI] [PubMed] [Google Scholar]
- 128. Aldunate M, Tyssen D, Johnson A, et al. Vaginal concentrations of lactic acid potently inactivate HIV. J Antimicrob Chemother 2013; 68:2015–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Petrova MI, van den Broek M, Balzarini J, Vanderleyden J, Lebeer S. Vaginal microbiota and its role in HIV transmission and infection. FEMS Microbiol Rev 2013; 37:762–92. [DOI] [PubMed] [Google Scholar]
- 130. Abdool Karim SS, Baxter C, Passmore JS, McKinnon LR, Williams BL. The genital tract and rectal microbiomes: their role in HIV susceptibility and prevention in women. J Int AIDS Soc 2019; 22:e25300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Nelson TM, Borgogna JL, Brotman RM, Ravel J, Walk ST, Yeoman CJ. Vaginal biogenic amines: biomarkers of bacterial vaginosis or precursors to vaginal dysbiosis? Front Physiol 2015; 6:253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Ziklo N, Huston WM, Taing K, Katouli M, Timms P. In vitro rescue of genital strains of Chlamydia trachomatis from interferon-γ and tryptophan depletion with indole-positive, but not indole-negative Prevotella spp. BMC Microbiol 2016; 16:286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Ziklo N, Huston WM, Hocking JS, Timms P. Chlamydia trachomatis genital tract infections: when host immune response and the microbiome collide. Trends Microbiol 2016; 24:750–65. [DOI] [PubMed] [Google Scholar]
- 134. Goytia M, Shafer WM. Polyamines can increase resistance of Neisseria gonorrhoeae to mediators of the innate human host defense. Infect Immun 2010; 78:3187–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Gong Z, Tang MM, Wu X, et al. Arginine- and polyamine-induced lactic acid resistance in Neisseria gonorrhoeae. PLoS One 2016; 11:e0147637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Ma B, France MT, Crabtree J, et al. A comprehensive non-redundant gene catalog reveals extensive within-community intraspecies diversity in the human vagina. Nat Commun 2020; 11:940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Muzny CA, Taylor CM, Swords WE, et al. An updated conceptual model on the pathogenesis of bacterial vaginosis. J Infect Dis 2019; 220:1399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Macklaim JM, Fernandes AD, Di Bella JM, Hammond JA, Reid G, Gloor GB. Comparative meta-RNA-seq of the vaginal microbiota and differential expression by Lactobacillus iners in health and dysbiosis. Microbiome 2013; 1:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Wylie KM, Wylie TN, Cahill AG, Macones GA, Tuuli MG, Stout MJ. The vaginal eukaryotic DNA virome and preterm birth. Am J Obstet Gynecol 2018; 219:189.e1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Happel A-U, Varsani A, Balle C, Passmore J-A, Jaspan H. The vaginal virome-balancing female genital tract bacteriome, mucosal immunity, and sexual and reproductive health outcomes? Viruses 2020; 12:832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Galiwango RM, Bagaya B, Mpendo J, et al. Protocol for a randomized clinical trial exploring the effect of antimicrobial agents on the penile microbiota, immunology and HIV susceptibility of Ugandan men. Trials 2019; 20:443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Klatt NR, Cheu R, Birse K, et al. Vaginal bacteria modify HIV tenofovir microbicide efficacy in African women. Science 2017; 356:938–45. [DOI] [PubMed] [Google Scholar]
- 143. Gatski M, Martin DH, Levison J, et al. The influence of bacterial vaginosis on the response to Trichomonas vaginalis treatment among HIV-infected women. Sex Transm Infect 2011; 87:205–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Bradshaw CS, Morton AN, Hocking J, et al. High recurrence rates of bacterial vaginosis over the course of 12 months after oral metronidazole therapy and factors associated with recurrence. J Infect Dis 2006; 193:1478–86. [DOI] [PubMed] [Google Scholar]
- 145. Sobel JD, Ferris D, Schwebke J, et al. Suppressive antibacterial therapy with 0.75% metronidazole vaginal gel to prevent recurrent bacterial vaginosis. Am J Obstet Gynecol 2006; 194:1283–9. [DOI] [PubMed] [Google Scholar]
- 146. Balkus JE, Manhart LE, Lee J, et al. Periodic presumptive treatment for vaginal infections may reduce the incidence of sexually transmitted bacterial infections. J Infect Dis 2016; 213:1932–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Balkus JE, Srinivasan S, Anzala O, et al. Impact of periodic presumptive treatment for bacterial vaginosis on the vaginal microbiome among women participating in the preventing vaginal infections trial. J Infect Dis 2017; 215:723–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Schwebke JR, Desmond R. A randomized trial of metronidazole in asymptomatic bacterial vaginosis to prevent the acquisition of sexually transmitted diseases. Am J Obstet Gynecol 2007; 196:517.e1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Schwebke JR, Lee JY, Lensing S, et al. Home screening for bacterial vaginosis to prevent sexually transmitted diseases. Clin Infect Dis 2016; 62:531–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Balkus JE, Carter KA, McClelland RS. Lessons from suppressive therapy and periodic presumptive treatment for bacterial vaginosis. Curr Infect Dis Rep 2019; 21:34. [DOI] [PubMed] [Google Scholar]
- 151. Joag V, Obila O, Gajer P, et al. Impact of standard bacterial vaginosis treatment on the genital microbiota, immune milieu, and ex vivo human immunodeficiency virus susceptibility. Clin Infect Dis 2019; 68:1675–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Cohen CR, Wierzbicki MR, French AL, et al. Randomized trial of lactin-V to prevent recurrence of bacterial vaginosis. N Engl J Med 2020; 382:1906–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Vrancken G, Gregory AC, Huys GRB, Faust K, Raes J. Synthetic ecology of the human gut microbiota. Nat Rev Microbiol 2019; 17:754–63. [DOI] [PubMed] [Google Scholar]
- 154. Lev-Sagie A, Goldman-Wohl D, Cohen Y, et al. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat Med 2019; 25:1500–4. [DOI] [PubMed] [Google Scholar]
- 155. Marrazzo JM, Dombrowski JC, Wierzbicki MR, et al. Safety and efficacy of a novel vaginal anti-infective, TOL-463, in the treatment of bacterial vaginosis and vulvovaginal candidiasis: a randomized, single-blind, phase 2, controlled trial. Clin Infect Dis 2019; 68:803–9. [DOI] [PMC free article] [PubMed] [Google Scholar]