Skip to main content
Journal of the International AIDS Society logoLink to Journal of the International AIDS Society
. 2019 May 29;22(5):e25300. doi: 10.1002/jia2.25300

The genital tract and rectal microbiomes: their role in HIV susceptibility and prevention in women

Salim S Abdool Karim 1,2,, Cheryl Baxter 1, Jo‐Ann S Passmore 1,3,4, Lyle R McKinnon 1,5,6, Brent L Williams 2,7
PMCID: PMC6541743  PMID: 31144462

Abstract

Introduction

Young women in sub‐Saharan Africa are disproportionately affected by HIV, accounting for 25% of all new infections in 2017. Several behavioural and biological factors are known to impact a young woman's vulnerability for acquiring HIV. One key, but lesser understood, biological factor impacting vulnerability is the vaginal microbiome. This review describes the vaginal microbiome and examines its alterations, its influence on HIV acquisition as well as the efficacy of HIV prevention technologies, the role of the rectal microbiome in HIV acquisition, advances in technologies to study the microbiome and some future research directions.

Discussion

Although the composition of each woman's vaginal microbiome is unique, a microbiome dominated by Lactobacillus species is generally associated with a “healthy” vagina. Disturbances in the vaginal microbiota, characterized by a shift from a low‐diversity, Lactobacillus‐dominant state to a high‐diversity non‐Lactobacillus‐dominant state, have been shown to be associated with a range of adverse reproductive health outcomes, including increasing the risk of genital inflammation and HIV acquisition. Gardnerella vaginalis and Prevotella bivia have been shown to contribute to both HIV risk and genital inflammation. In addition to impacting HIV risk, the composition of the vaginal microbiome affects the vaginal concentrations of some antiretroviral drugs, particularly those administered intravaginally, and thereby their efficacy as pre‐exposure prophylaxis (PrEP) for HIV prevention. Although the role of rectal microbiota in HIV acquisition in women is less well understood, the composition of this compartment's microbiome, particularly the presence of species of bacteria from the Prevotellaceae family likely contribute to HIV acquisition. Advances in technologies have facilitated the study of the genital microbiome's structure and function. While next‐generation sequencing advanced knowledge of the diversity and complexity of the vaginal microbiome, the emerging field of metaproteomics, which provides important information on vaginal bacterial community structure, diversity and function, is further shedding light on functionality of the vaginal microbiome and its relationship with bacterial vaginosis (BV), as well as antiretroviral PrEP efficacy.

Conclusions

A better understanding of the composition, structure and function of the microbiome is needed to identify opportunities to alter the vaginal microbiome and prevent BV and reduce the risk of HIV acquisition.

Keywords: HIV prevention, women, vaginal microbiome, rectal microbiome, genital inflammation, tenofovir gel


Abbreviations

BV

bacterial vaginosis

Cpn60

chaperonin‐60

CST

community state type

HEPN

highly exposed seronegative

HIV

human immunodeficiency virus

HMP

Human Microbiome Project

HPV

human papillomavirus

IL

interleukin

IP

inducible protein

LPS

lipopolysaccharide

MCP

monocyte chemotractant protein

MIP

macrophage inflammatory protein

MSM

men who have sex with men

NEC

necrotizing endocolitis

PrEP

pre‐exposure prophylaxis

PRR

prevalence risk ratio

SIV

simian immunodeficiency virus

SMS

Shotgun metagenomic sequencing

STI

sexually transmitted infections

TLR

toll‐like receptor

1. Introduction

With 37 million people living with human immunodeficiency virus (HIV) and 1.8 million new infections in 2017, HIV remains a significant global public health problem. Sub‐Saharan Africa bears a disproportionate HIV burden and accounts for 70% of all people living with HIV (PLHIV) 1. In this region, adolescent girls and young women (aged 15 to 24 years) are particularly vulnerable and, despite representing just 10% of the population, accounted for 25% of all new HIV infections in 2017. A recent report from a rural South African community illustrated the enormity of this problem where HIV prevalence was alarmingly high in women, reaching 40.1% in those 23 to 24 years and a staggering 71.7% in those aged 35 to 36 years 2. A complex interplay of biology, gender‐power disparities, socio‐economic and behavioural factors impacts vulnerability in women in Africa 3.

The female genital tract microbiome, which is the main site of infection for women, is a key, but lesser understood, biological factor affecting HIV susceptibility. In this review, we examine how disturbances in the female genital tract bacterial microbiome influence HIV acquisition and HIV prevention, describe the role of the rectal microbiome in HIV acquisition, outline advances in technologies to study the microbiome and postulate some future research directions in this field.

1.1. The human microbiome

For human hosts, the term “microbiome” includes all the genetic material of all the microbes (bacteria, fungi, protozoa and viruses) that exist in and on the human body. Trillions of microbial cells colonize the human skin and various mucosal niches 4, and are critically important for maintaining health. Disturbances in the microbiota, that is, all the microorganisms, in these various microbial niches have been linked to an array of adverse health outcomes, including altered gut microbiomes associated with inflammatory bowel disease 5, obesity 6, autoimmunity and heart disease 7, 8. Disturbances in the vaginal microbiome have been associated with reproductive complications (such as pelvic inflammatory disease, infertility, premature delivery, mid‐trimester pregnancy loss, amniotic fluid infection or low for gestational age infants, postpartum endometritis and gynaecologic postoperative infections 9. Transfer of gut bacteria can transmit phenotypes such as obesity and malnutrition to another animal both within and between species (for humans and mice) 10, 11, opening avenues for potential interventions. Transferring beneficial gut bacteria through faecal transplants has markedly improved the treatment of Clostridium difficile by improving the gut microbial environment 12.

In this review, we focus specifically on the female genital tract and rectal microbiomes and their association with HIV acquisition.

1.2. The female genital tract microbiome

The female genital tract microbiome comprises bacteria, protozoa, viruses and fungi inhabiting the human vagina, which may promote health (e.g. bacteria – Lactobacilli) or disease (e.g. bacteria – Chlamydia trachomatis and Neisseria gonorrhoeae; protozoa – Trichomonas vaginalis; viruses – Human papillomaviruses and Herpes simplex viruses; and fungi – Candida spp.). Studies 13, 14 examining whether there are differences between the cervical and vaginal microbiomes have revealed a high level of concordance in microbial diversity between the cervix and vagina. The composition of the female genital tract microbiome is unique to each woman and is probably established early on in life through exposure to key maternal microorganisms during birth 15, 16. Menarche and sexual debut also have an impact on the female genital tract microbiome, but their role has not been clearly characterized. While not fully understood, the microbial composition fluctuates naturally over time 16, particularly during hormonal shifts associated with puberty and menopause 17, 18 and through the menstrual cycle 17, 19.

In addition, several environmental factors such as hormonal contraceptives; sexual activity (including the number of partners and semen, as well as lubricants), hygiene practices, antibiotics and the composition of the gastrointestinal microbiota (transferred from the nearby rectum) can all influence the composition of the female genital tract microbiome 20, 21, 22, 23, 24, although it should be noted these data are not entirely consistent.

1.2.1. Eubiotic microbiota

The female genital tract microbiome of healthy women asymptomatic for vaginal dysbiosis is usually dominated by one or two species of Lactobacillus (including L. crispatus, L. iners, L. jensenii, L. mucosae and L. gasseri). Lactobacilli spp. are thought to benefit the host by producing lactic acid (a potent, broad‐spectrum bactericide and virucide) and hydrogen peroxide, which lowers the vaginal pH (<4.5) and promotes the production of bacteriocins that reduce colonization by other common pathogenic microorganisms 25, 26, 27, including HIV and other sexually transmitted infections (STIs) 28, 29. However, not all Lactobacillus species contribute equally to the stability of the normal vaginal microflora. A study among pregnant women has shown that L. gasseri and/or L. iners may predispose the occurrence of abnormal vaginal microflora while presence of L. crispatus promotes the stability of the normal vaginal microflora 30.

1.2.2. Dysbiotic microbiota

While it is generally thought that Lactobacillus‐dominant vaginal communities represent states of “health,” it is less clear whether all non‐Lactobacillus dominant communities are necessarily “unhealthy.” Several studies have highlighted racial and ethnic differences in healthy asymptomatic women 31, 32, 33. Generally, women of European and Asian ancestry are more likely to have a microbiota dominated by Lactobacillus while women of African or Hispanic descent are more likely to have non‐Lactobacillus‐dominated microbiota 34, 35, 36, 37. Up to 40% of apparently healthy women, with no clinical evidence of symptoms or discharge, have vaginal communities that lack appreciable numbers of Lactobacillus and instead have vaginal microbiomes that are dominated by a variety of other anaerobic bacteria 33, 34, 35, 38. This type of vaginal microbiome is common, especially in black and Hispanic women, and it is unclear whether this should be considered a “normal” or a dysbiotic/diseased state 38. The underlying factors determining this apparent tolerance of a non‐Lactobacillus‐dominated microbiota remain obscure. One hypothesis is that early‐life exposure to this type of vaginal microbiota could foster immunological tolerance 39. Other possibilities may relate to genetic variation between individuals and among ethnic populations, especially polymorphisms in cytokine, innate immunity and hormone‐response genes that could impact the threshold for immunological responses to bacteria in the vaginal tract 40, 41.

Furthermore, some anaerobes, like Atopobium, Megasphaera and Leptotrichia 35, 42, 43, are able to produce lactic acid and thus may be metabolically similar to species of Lactobacillus in their ability to lower vaginal pH 9, 16, 35, 38. However, it should be noted that these other lactic acid producers are frequently associated with bacterial vaginosis (BV) and high diversity vaginal bacterial communities, wherein Atopobium, Megasphaera and Leptotrichia are typically present at low relative abundance compared to Lactobacillus dominated vaginal bacterial communities, wherein Lactobacillus may account for up to 99% of the total bacterial relative abundance 44, 45, 46. Thus, the contribution of these relatively low‐abundance microbes to total lactic acid production in the vaginal tract may be minimal compared to vaginal communities that are dominated by Lactobacillus. In addition, their co‐occurrence with diverse anaerobes may counteract any pH lowering effects as some bacteria, including the common urogenital pathogen, Neisseria gonorrhoeae, have the capacity to take up lactic acid and utilize it as a carbon source, thus depleting it from their environments 47, and BV‐type microbial communities are associated with production of polyamines such as putrescine, cadaverine and trimethylamine, as well as ammonia which can counteract the pH lowering effects of lactic acid 48.

To simplify measurement of the female genital tract microbiome, several different algorithms have been applied to stratify women based on vaginal bacterial composition, by clustering them into various numbers of distinct Community State Types (CSTs, or vagitypes) 34, 38, 49, 50, 51, 52, 53, similar to enterotype clusters reported for the human gut microbiome 54. For example, the classification by Ravel and colleagues 38 proposed five CSTs where CST I, II, III and V are composed primarily of Lactobacillus species including L. crispatus, L. gasseri, L. iners or L. jensenii respectively while CST IV contains lower proportions of Lactobacillus species and a higher diversity of several other anaerobic organisms 38. CST IV was subsequently further divided into IVA (consisting primarily of Bifidobacterium spp., Dialister spp., Streptococcus spp. and Bacteroides spp.) and IVB (consisting of Atopobium spp., Gardnerella spp., Mobiluncus spp., Paryimonas spp., Prevotella spp., Megasphaera spp. and several other anaerobic bacteria) 55. However, methodological differences, assessment of different populations of women and limitations of sequencing short regions of marker genes for taxonomic discrimination have contributed to the lack of a universal definition of CSTs. Despite their common usage and utility, it remains unclear whether this categorization adequately represents biologically and clinically relevant entities in the vaginal environment.

1.3. Dysbiosis of the vaginal microbiome, with a focus on bacterial vaginosis

An imbalance in the vaginal microbiota, characterized by a shift from a low‐diversity, Lactobacillus‐dominant state to a high‐diversity state in which the vagina is colonized by a wide range of strict and facultative anaerobes 40, 56, 57, 58, 59 and vaginal pH ≥4.5 57, 58, 60, is associated with a clinical condition referred to as bacterial vaginosis (BV). Although BV is not considered to be an infection (STI), it is known to be associated with condomless sexual contact 40, 61. Despite BV being common in reproductive‐age women, it remains an enigma in women's health. More than 50% of women with BV are asymptomatic 62, 63, highlighting the complexity of referring to BV as an illness. The complexity surrounding BV arises, in part, from a nebulous definition of the disorder and subjective diagnostic criteria 43.

Nugent scoring is most commonly used to diagnose BV in a research setting, based on the presence or absence of Lactobacilli rods by Gram staining 64. Alternatively, BV is diagnosed clinically based on the presence of three of the four Amsel criteria: (1) pH of vaginal fluid > 4.5, (2) presence of clue cells, (3) vaginal discharge and (4) malodour 62. BV‐associated symptoms and categories of vaginal dysbiosis (i.e. Lactobacillus dominant vs. non‐Lactobacillus dominant); however, may lack the requisite granularity to enhance our understanding of the complex aetiology of BV, discriminate important aspects of bacterial community structure and function, and discern host‐microbiome interactions that may serve to facilitate or inhibit HIV transmission.

Women with BV typically have a spectrum of anaerobes, including Gram‐positive anaerobic bacteria like Gardnerella vaginalis, Atopobium vaginae, Eggerthella spp., Peptoniphilus spp. and BV‐associated bacteria (BVAB1‐3, Clostridia‐like bacteria), anaerobic Gram‐negative bacteria like Mobiluncus spp., Prevotella spp., Fusobacterium nucleatum and Megasphaera type 1, and bacteria without cell walls, like Mycoplasma hominis and Ureaplasma spp 38, 45, 57, 61. Of these, biofilm‐producing G. vaginalis is thought to play a pivotal role in the initiation and persistence of BV 65, 66.

In one of the earliest studies seeking to understand the aetiology of BV, Criswell et al. 67. inoculated pure cultures of G. vaginalis into the vaginas of 13 healthy women. Only one of the 13 women developed BV. Separately, he inoculated 15 women with vaginal fluid from women with BV; and 11 of the 15 developed BV 61, 67. Subsequently, a wide range of host and environmental factors have been implicated in the aetiology of BV, including cigarette smoking 68, early age of sexual intercourse 69, oral sex 23, new or multiple sexual partners 21, menses and having intercourse while menstruating 19, and frequent douching (more than once a week) 70. However, several of these factors may be confounded, as many of the socio‐demographic factors that increase the risk of BV are more common in black women; or it may be that the symptoms of BV (such as malodour and discharge) cause women to douche rather than that douching has a direct effect on vaginal microbes 32, 71.

Sexual partners are thought to be a reservoir for BV‐associated bacteria 61. G. vaginalis, A. vaginae, BVAB1 and Megasphaera sp. type 1 have all been detected in the male partners of women with BV, with the coronal sulcus suggested to have the highest concentrations 72. In addition, semen was found to harbour high concentrations of bacteria, with some of the most abundant genera being Prevotella, Streptococcus, Staphylococcus as well as Lactobacilli 73. The penile skin and urethral microbiota of male partners of women with BV were more similar to the vaginal microbiota of their female partners than the vaginal microbiota of non‐partner women with BV 72. However, a recent meta‐analysis of seven placebo‐controlled trials, that included more than 1700 participants, showed that treating male partners with antibiotics does not significantly improve BV treatment outcomes or reduce recurrence in female partners 74. A limitation of these early trials were that test of cure was not conducted in the male partners, who were only treated with oral antibiotics, and the primary outcome of the trials was clinical cure (Amsel criteria) in women within one to two months of partner treatment. More recently, Plummer et al. 75 revisited this approach, using 16S rRNA gene sequencing and combined oral metronidazole and topical penile clindamycin treatment of male partners of women with BV, to show that a more homogeneous microbiota can be restored in women with this combined treatment approach, with a reduction in BV‐associated bacterial species. Furthermore, partners had an immediate effect on the penile cutaneous microbiota. Since male partner‐derived BV‐associated bacteria can colonize the lower FGT, it is probable that this microbiome sharing does modulate risk for HIV infection in women by causing BV, in addition to influencing topical PrEP efficacy.

A different school of thought is that the overgrowth of pathogens characteristic of BV has to be preceded by a major disturbance in the vaginal Lactobacillus population, such as specific targeting by bacteriophages 76, 77, 78. Bacteriophages targeting Lactobacilli have been isolated from various environmental sources, such as dairy products 79 and the human gastrointestinal tract 80. In addition, temperate phages are evidence in sequences from a wide range of human vaginal Lactobacilli spp. that are capable of infecting other Lactobacilli isolates from the same and different women 81, 82. In trying to understand major drivers of the shift from microbial health to dysbiosis in the vagina, significantly more work is required to better understand what BV is, how other constituents of the microbiome interact with Lactobacilli, and how to prevent and durably treat this common condition.

1.4. Dysbiosis of the female genital tract microbiome and the risk of HIV

Disturbances in the vaginal microbiota, associated with BV, increase genital inflammation and HIV target cell activation, likely through inflammatory cytokine induction 45, 83, 84. Raised pre‐infection genital concentrations of inflammatory cytokines (macrophage inflammatory protein (MIP)‐1α and MIP‐1β) were present in women who acquired HIV infection compared to women who remained uninfected 83. These inflammatory changes were associated with elevated levels of highly activated T cells in the genital mucosa 45, 84 and decreased efficacy of the antiretroviral drug tenofovir, which is a reverse transcriptase inhibitor that was assessed for its effectiveness in preventing HIV infections 85. In rhesus macaques, the establishment of a productive simian immunodeficiency virus (SIV) infection in the genital mucosa required local chemokines, which facilitated recruitment of CD4+ T cells to the site of infection, and enabled SIV expansion and establishment of infection 86. Li et al. 86 further showed that suppression of inflammation prior to SIV inoculation prevented SIV infection. In addition to recruiting target cells to the site of transmission, there is evidence showing that pro‐inflammatory cytokines facilitate HIV transmission by reducing the integrity of the epithelial barrier in the female genital tract and directly promoting HIV replication through NF‐kB activation 87, 88, 89.

While inflammatory cytokines are induced during BV, several chemokines are down‐regulated, including those associated with HIV risk (interleukin (IL)‐8, interferon‐γ inducible protein (IP)‐10 and monocyte chemotractant protein (MCP)‐1) 83. The reasons for suppressed chemokine expression in women who have BV remain unclear 40. Since BV is a complex polymicrobial condition, identifying precisely which microbes are associated with mucosal inflammation and barrier dysfunction is difficult. It is likely that this results from multiple pathogenic anaerobes 40.

Several recent studies have suggested that G. vaginalis and P. bivia contribute to both HIV risk and genital inflammation 45, 90, 91. Several virulence factors have been identified in both of these BV‐associated microbes. G. vaginalis produces several classes of cytolysin, including vaginolysin, which are capable of activating the protein kinase pathway in vaginal epithelial cells, inducing cell death 92. Gardnerella spp. also commonly produce sialidase, prolidase and putrescine, which degrade mucins, allow better microbial attachment and biofilm formation, and may contribute to epithelial sloughing during BV 93. Similarly, Prevotella spp., including P. bivia, produce collagenases, fibrinolysins, sialidases and prolidases 94. Metagenomic analysis of vaginal 16S sequences from women with BV suggested that lipopolysaccharide (LPS) from P. bivia was the strongest predictor of both genital inflammation and HIV risk in women 91. LPS signals through toll‐like receptor (TLR)‐4 and CD14, which are expressed by monocytes and macrophages, as well as epithelial cells in the cervix, vagina, fallopian tube and endometrium 95. TLR4 expression has been shown to decline progressively along the genital tract, being highest in the fallopian tubes and endometrium, and lowest towards the vaginal epithelium 96. Furthermore, some studies have suggested that the expression of TLR4 in the endometrium is hormonally regulated; being significantly higher during the secretory phase compared to other phases of the menstrual cycle 97. LPS binds to TLR‐4 and CD14 to activate the NF‐Kb cytokine pathway 95. P. bivia is a common constituent of BV, and known to produce high concentrations of LPS in vaginal washes 98. In an in vitro host cell cytotoxicity model using normal dopamine neuron cells, LPS from vaginal P. bivia induced cytotoxicity, although this needs to be confirmed in vaginal or cervical cells. Furthermore, in a study of inflammation caused by a periodontal Prevotella spp., P. nigrescens induced NF‐Kb inflammatory pathways involving nitric oxide in a murine macrophage model 99. It is therefore possible that this anaerobic pathobiont contributes substantially to genital inflammation, which may influence barrier disruption and increased HIV risk in women with BV.

1.5. Maintenance of vaginal health

Maintaining or restoring vaginal health is thought to play an important role in protecting women from reproductive complications and acquisition of STIs, including HIV and human papillomavirus (HPV) 45, 60, 70, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111.

The most common treatment for BV includes either oral or topical metronidazole, although more than half of those receiving antibiotic treatment have recurrent BV within a year 112. More than 50 therapeutically bioactive compounds, including metronidazole, are known to undergo direct microbial modification in the gastrointestinal tract 113, 114, thereby altering their chemical structure and modifying their half‐life, bio‐availabilities and biological effects. Gut microbes predominantly use hydrolytic and/or reducing reactions to metabolize xenobiotics 115, and many of the enzymes associated with xenobiotic metabolism (including hydrolases, lyases, oxidoreductases and transferases) are common among gut microbial sequences 114. Although some of these modifications can be useful (conversion of inactive pro‐drugs into their active forms), many of these modifications lead to detoxification and/or inactivation of the compounds 113. Changes in metabolism of various drugs in germ‐free animals or following antibiotic treatment have confirmed the role of gut microbes in xenobiotic processing 115, 116.

Higher oestradiol levels have been shown to favour the dominance and stability of Lactobacilli spp. in the lower reproductive tract through the oestrogen‐mediated increase in the thickness and glycogen content of the vaginal epithelia, which peaks mid‐cycle 117, 118. Bacterial communities were shown to be more stable when oestradiol levels are at their highest, and the greatest change in the complexity of microbial communities was found at the onset of menstruation 19, 119, 120. A systematic review and meta‐analysis showed that some hormonal contraceptives reduce the risk of BV, through hormonal stabilization of vaginal microbial communities, arguing that a hormonal intervention could be useful in BV prevention 121, 122.

Pregnancy is an interesting endocrinological and immunological phase of life for women, where levels of both oestrogen and progesterone rise continually throughout pregnancy. Despite changing physiology, hormones and mucosal tolerance during pregnancy, several studies have shown that the vaginal community composition during gestation is relatively stable 16, 44, 123, with higher abundance of Lactobacilli spp. than non‐pregnant women, like L. crispatus, L. gasseri and L. jensenii, and lower relative abundance of BV‐associated organisms, like Prevotella, Sneathia, Gardnerella and Mobiluncus 44. Romero et al. (2014) speculated that the higher vaginal community stability during pregnancy than non‐pregnancy may be a consequence of the physiological state of pregnancy 44. Similar to pregnancy, stability of vaginal microbial communities is highest around ovulation in non‐pregnant women 16, when oestrogen concentrations are high the vaginal epithelium mature, and epithelial glycogen stores as a carbon source for Lactobacillus spp. are higher 124. Pregnancy may therefore reflect an extended oestrogenic state, in the absence of menstruation, that favours Lactobacilli microbial stability.

Condom use can prevent BV. Women whose partners always used condoms were fivefold less likely to have persistent or recurrent BV 125. In contrast, women reporting less frequent condom use were less likely to have or sustain Lactobacilli‐dominant vaginal microbiota 126. Medical male circumcision protected women against BV. The female partners of men who participated in a randomized trial of circumcision to prevent HIV acquisition in Uganda were significantly less likely to develop BV than partners of uncircumcised men 61, 127. These findings suggest that male partners are a likely source of the organisms associated with BV, and transfer of these during condomless sex, particularly from male partners who have intact foreskins, results in displacement of commensal vaginal Lactobacilli.

Lactobacilli‐containing probiotics have had varying success in preventing BV recurrence, with a recent meta‐analysis including 1304 women from 12 RCTs, showing that probiotic supplementation significantly improved BV cure rates 128, although this was not significantly better than metronidazole treatment alone 129. However, long‐term colonization with the probiotic has proved difficult to achieve, although few products are explicitly for vaginal conditions and even fewer contain common vaginal Lactobacilli spp 130, 131, 132. It is likely that the same factors that caused disappearance of endogenous vaginal Lactobacilli in the first place may limit the colonizing success of introduced probiotic species.

1.6. The role of the rectal microbiome in HIV acquisition

In both men and women, receptive anal intercourse is associated with higher rates of HIV acquisition. The rectal immune environment is influenced by the gut microbiome, which a growing body of literature suggests is an important determinant of overall immune function 133, 134. The composition of the human gut and rectal microbiome change with age, influenced by a wide range of factors including diet, antibiotic use, gender and geographical location 135.

The gut has been a major focus of HIV research for over two decades, after initial reports of HIV‐related enteropathy. This was followed by demonstration of rapid gut CD4 depletion in both HIV‐infected humans and SIV‐infected non‐human primates in the days/weeks following infection 136, 137. Moreover, the resulting damage caused to the gut during this early phase of infection has been implicated as a driver of pathogenesis, specifically by causing translocation of microbial components into the blood stream and causing inflammation 138. Several studies have suggested that HIV alters the microbiome 139, 140, 141, 142. In addition to differences in species composition, altered bacterial metabolic activity has been noted in HIV infection 143, 144. However, recent studies have suggested that at least some of the differences between the gut microbiota of HIV‐positive and HIV‐negative individuals may be linked to sexual preference 145, 146, leaving an open question as to how HIV infection impacts the microbial composition of the gut, and in turn how this impacts its interaction with the host immune system (also greatly impacted by HIV). An even larger open question is the reverse relationship – whether the gut microbiota and associated immune environment of the rectum influence HIV susceptibility.

Comparatively, a greater role for commensal bacteria playing a role in HIV acquisition has been better described for the female genital tract 90, 111 than in the gut. While diversity in the vagina is likely a result of the loss of the physiologically critical Lactobaccillus spp., as discussed above, in the gut a diverse microflora is believed to be advantageous for improved nutrient absorption. While the extent to which the gut microbiome may be implicated in rectal HIV acquisition is unknown, the gut microbiome plays a critical regulatory role in defining the rectal immune environment by signalling through pattern recognition receptors 147, setting up what has been referred to as a “mucosal firewall” and controlling host‐advantageous communication with the external environment. The gut microbiome is also associated with mucosal Th17 and Treg frequencies 148, cell subsets that play important roles in HIV transmission 149, 150.

The role of rectal microbiota in HIV acquisition has been explored in recent animal and human studies. A study in non‐human primates demonstrated that differences in SHIV susceptibility was due primarily to gut microbial differences 151, as the more susceptible group of animals had higher levels of immune activation that were linked to lower Bacteroides and Firmicutes, and higher proportions of Prevotella spp. In a human study, condomless rectal sex was found to have a substantial inflammatory effect on the rectal immune environment, including on composition of the microbiota inflammation‐associated bacteria from the Prevotellaceae family 146. This microbial profile was also associated with increases in Th17 cells, pro‐inflammatory cytokines, and a gene expression signature of wound injury. These findings echo findings of the important inflammatory role for Prevotella in the female genital tract, suggesting that this bacterial species in the microbiome of both compartments may be contributing to HIV acquisition.

1.7. The female genital tract microbiome and its impact on antiretroviral drug concentrations

The composition of the vaginal microbiome has been shown to have a direct impact on the efficacy of some antiretroviral drugs that are used as pre‐exposure prophylaxis (PrEP) for HIV prevention, particularly those administered intravaginally. In one study, the effectiveness of topical tenofovir gel against HIV was diminished substantially in women with BV‐associated bacteria compared to women with a Lactobacillus‐dominated microbiota 90. In this study, tenofovir concentrations were significantly depleted in vitro (by >50%) within four hours in cultures with G. vaginalis (the bacterial isolates that were most common in women with non‐Lactobacillus dominated microbiota) compared to only marginal changes in cultures with Lactobacillus species. In addition, the tenofovir metabolite, adenine, increased progressively in Gardnerella cultures as tenofovir concentrations declined, indicating that this BV‐associated organism was metabolizing tenofovir 90. The metabolism of tenofovir by G. vaginalis has also been shown in a recent study 152 that found that tenofovir concentrations were lower in both cervicovaginal fluid and plasma of women with BV‐associated G. vaginalis and A. vaginae after applying tenofovir gel or tenofovir film for a week. Dapivirine, the antiretroviral being used in intravaginal rings for HIV prevention, has also been shown to be impacted by BV‐associated bacteria, although the mechanism differs from tenofovir 153. Unlike tenofovir, dapivirine does not require intracellular modifications and its pharmacokinetics are therefore not disrupted by BV organisms. Instead, Gardnerella spp. appear to bind dapivirine irreversibly, thereby reducing drug concentrations 153, 154. Recent studies have found that tenofovir alefenamide, a pro‐drug of tenofovir, was not metabolized by BV‐associated bacteria suggesting it may be a more appropriate antiretroviral drug for PrEP in women with BV 153, 154. The composition of the vaginal microbiome; however, does not appear to impact on the effectiveness of oral tenofovir‐containing PrEP 155.

1.8. Changing technologies to study the structure and function of the genital microbiome

The application of –omic technologies to assess the vaginal and rectal microbiome and host responses to dysbiotic states have the potential to provide a greater systems‐level understanding of states of health and disease that influence the risk for STIs such as HIV, particularly when integrated with measures of host physiology, genetics and environmental and behavioural factors. Such a systems approach may be particularly important in the context of HIV susceptibility wherein microbial and host factors and their inextricable and complex interactions at mucosal sites may promote conditions that either increase or reduce the risk of HIV acquisition. As the application of newly evolving technologies expands the ability to study microbiome‐host interactions in the vaginal ecosystem, these tools can be applied more broadly to other mucosal ecosystems, including the rectal mucosal environment.

1.8.1. Amplicon‐based sequencing approaches

The advent of cultivation‐independent, next‐generation sequencing has dramatically advanced our knowledge of the diversity and complexity of the human microbiome. To date most studies of the vaginal microbiome have employed high throughput sequencing targeting bacterial marker genes, such as the 16S rRNA or chaperonin‐60 (cpn60) genes 25, 156, 157, 158. In addition to providing more comprehensive surveys of vaginal microbial communities, 16S rRNA gene sequencing has led to the identification of previously unrecognized BV‐associated bacteria in the order Clostridiales; BVAB1, BVAB2 and BVAB3 (now isolated and classified as Mageeibacillus indolicus) 159, 160, 161, and cpn60 sequencing has been utilized to resolve different G. vaginalis subgroups which may have variable clinical and pathological relevance for BV 162, 163, 164, 165, 166.

Although compositional analyses using marker gene sequencing have led to important insights into the complexity of the vaginal microbiota as it relates to BV, vaginal inflammation and HIV acquisition 49, 110, 111, 167, it has several important limitations that need to be considered, such as the inability to provide fine scale taxonomic resolution (species‐ and strain‐level), that these are subject to primer biases, and that they cannot provide exact information about the metabolic functional capacity of bacterial communities.

1.8.2. Shotgun metagenomic sequencing

Shotgun metagenomic sequencing (SMS), while more costly and computationally demanding, can overcome many of the limitations of amplicon‐based sequencing by improving taxonomic classification. SMS can also identify other important components of the vaginal microbiome (such as viruses, fungi, archaea and protozoa) and thereby shed light on the broader relationships with the vaginal microbiome that cannot be appreciated using bacterial amplicon‐based approaches alone. Despite these advantages, only a few published vaginal SMS datasets are currently available; one was derived from a subset of 113 U.S. adult females as part of the Human Microbiome Project (HMP) and another from a recent longitudinal study of 10 pregnant women in the U.S 52, 168. Analyses of HMP vaginal SMS data revealed an association between vaginal pH and metabolic diversity. However, this U.S. cohort may be inadequate to assess states of vaginal health and disease as very few women had vaginal pH >4.5 169. Anahtar et al. applied SMS to evaluate functional differences in the female genital tract microbiome of 12 black South African women from the FRESH cohort (six women with high vs. six with low genital inflammation) and found that the pathway for lipopolysaccharide biosynthesis was enriched in the vaginal metagenomes of women with high inflammation 45. Additionally, in 180 women from the FRESH cohort, Gosmann et al. assessed the female genital tract virome using SMS with a virus enrichment protocol, identifying alphapapillomaviruses, Anelloviridae and Caudovirales as predominant viral taxa. However, no relationship between the virome and bacterial communities based on amplicon‐sequencing were identified 111. Thus, there is a paucity of SMS datasets for the vaginal microbiome and a need for and greater appreciation of differences between women from different geographic populations. Assessment of vaginal microbial composition based on SMS is superior to amplicon sequencing in that it can provide species and even strain level resolution of bacterial taxa using marker gene approaches, such as MetaPhlAn2 52, alignment of raw sequences to whole genome databases 170, or genome assembly‐based approaches 171, 172. However, such analyses are limited to bacterial taxa and strains for which full bacterial genomes have been assembled and deposited in databases. Thus, genomes of certain important vaginal bacterial species and strains that may have clinical relevance are missing from databases for MetaPhlAn2 and whole genome databases (i.e. Sneathia spp., BVAB1, BVAB2, and likely others), and such omissions can have dramatic effects in skewing abundance profiles and misclassifying samples to CSTs, especially in samples where such bacterial taxa may be dominant. In this regard, whole‐genome sequencing and comparative genomics of vaginal bacterial isolates could advance the accurate assessment of the vaginal microbial community structure and function, as has been undertaken for select vaginal isolates 66, 173, 174, 175, 176. One of the greatest strengths of SMS is the gene‐ and function‐level information it provides on the microbiome. The functional metabolic capacity of microbial communities could help to provide a clearer picture of vaginal health. Given that some BV‐associated taxa are also capable of lactic acid production, microbial community function could be a greater predictor of vaginal health that is dependent on context (i.e. HIV risk or susceptibility to other STIs as opposed to BV symptoms). SMS datasets in large cohorts would help to distinguish such context‐specific definitions of health and disease.

1.8.3. Metatranscriptomics and metaproteomics

Complementary ‐omic technologies aimed at evaluating the functional capacity of the microbiome, including metatranscriptomics and metaproteomics, offer further opportunities to expand our understanding of gene transcriptional activity and protein expression respectively in complex microbial communities inhabiting the human body. To date, metatranscriptomics with RNA‐seq has only been exploited in a handful of small studies of the vaginal microbiome 58, 177, 178. Vaginal bacterial metatranscriptome comparison between two women with BV and two healthy individuals indicated that in BV, L. iners increases transcription of cholesterol‐dependent cytolysin, genes involved in glycerol metabolism, and CRISPR‐related genes 58. Comparing the vaginal bacterial metatranscriptome in BV between metronidazole treatment responders and non‐responders, found that increased transcription of CRISPR‐Cas genes in G. vaginalis in non‐responders may mitigate treatment resistance 178. Metatranscriptomic analyses of cultured vaginal bacteria under different growth conditions may also provide important information. In vitro growth of G. vaginalis isolates in planktonic culture compared to biofilm growth has revealed extensive differences in the metatranscriptome that may be important for biofilm persistence and virulence 179. The emerging field of metaproteomics, studying bacterial proteins by mass spectrometry, is further shedding light on functionality of the vaginal microbiome and its relationship with BV, as well as antiretroviral drug efficacy, and has recently been extensively reviewed 180. Metaproteomics can provide important information on vaginal bacterial community structure, diversity and function and can further be applied to cultured isolates to better understand the protein repertoire expressed by individual vaginal bacteria. More recently, a “cell shaving proteomic” approach was used to identify 261 cell surface‐associated proteins of G. vaginalis in culture 181. This technology has several advantages as the metaproteome reflects the actual protein output, which may diverge from measures of bacterial community structure defined by amplicon sequencing, gene content (SMS) or metatranscriptomes 180, 182, 183.

1.8.4. Metabolomics

The human microbiome, including the vaginal ecosystem, executes a wide range of metabolic activities, producing a plethora of metabolites in the process, which may underlie important microbe‐host driven mechanisms distinguishing phenotypic traits. Advancements in both targeted and comprehensive untargeted metabolomics have already provided evidence for divergent metabolic activity associated with vaginal dysbiotic states, including abnormalities in sugar compound, short chain fatty acid, lipid, biogenic amine and amino acid metabolism 48, 184, 185, 186, 187, 188, 189, 190. However, little is known about vaginal metabolic profile disturbances in the context of STIs 191 or metabolite biomarkers that may be associated with infection risk 192, 193. Studies assessing vaginal metabolic profiles in the context of BV treatment (such as rifaximin) efficacy, can inform the development of effective clinical treatments 194. Greater appreciation of the impacts of individual bacterial metabolites on vaginal physiology could provide important mechanistic insights into states of vaginal health and disease. It should further be noted that the expansive metabolic capacity of the microbiome can impact drug efficacy and safety, as some bacteria of the microbiome can directly metabolize xenobiotic 195, 196, 197 or impact drug responses through immune‐mediated mechanisms, as suggested for immune checkpoint inhibitors used in cancer treatment 197, 198, 199, 200.

1.9. Suggestions for future research

With the wide range of available –omic technologies, the microbiome research field stands poised to address many of the questions that have led to an inadequate definition of vaginal health and the factors mediating STI outcomes. However, a better functional definition of BV and vaginal states that influence STI risk, in addition to microbial factors, will also require substantial effort to understand human host intrinsic (hormonal fluctuations, genetic polymorphisms, inflammatory responses), behavioural (sexual activity, contraception usage, vaginal cleansing practices, stress, physical activity, smoking, diet) and sociodemographic (age, ethnicity, geographic location) factors that may influence interactions at the host‐microbe interface. Given the temporal instability of the vaginal microbiome 16, 201, longitudinal, rather than cross‐sectional, study designs will further help to elucidate factors mediating transitions between healthy and unhealthy states. Finally, the application of multi‐omic approaches to study the microbiome and the host, leading to the acquisition of detailed study metadata will require novel approaches for integrating and interpreting such high dimensional datasets. Network modelling and machine learning approaches that can contend with such heterogeneous, complex datasets are being used increasingly in biology 202, 203, 204, 205. Some such analytical approaches, like MIMOSA, which facilitates integration of bacterial community data with metabolomic data, have been developed specifically with a focus on improving our understanding of the microbiome 190. Undoubtedly, additional bioinformatic tools are already in development that will further our understanding of the complex interactions between the microbiome and human factors in multiplex, fluctuating biological environments such as the vaginal tract.

2. Conclusions

In conclusion, the vaginal microbiome is a complex environment that is continually shifting. Imbalances in the vaginal microbiota are associated with a range of adverse reproductive health outcomes, including increasing the risk of genital inflammation and HIV acquisition. While there does not appear to be any impact of BV on oral PrEP effectiveness, the finding that BV‐associated organisms actively undermine HIV prevention modalities by degrading antiretroviral drugs, there is an urgent need to consider a more generalized role of these microbes in modifying the effect of biomedical HIV prevention tools, and whether the effect extends to long‐acting or systemic PREP strategies. However, maintaining vaginal health with antibiotics and probiotics has proved challenging. A better understanding of the composition, structure and function of the microbiome through advances in sequencing technologies may enable us to develop new strategies to make the vaginal microbiome healthy and prevent the occurrence of BV and reduce the risk of HIV acquisition.

Competing interests

The authors declare no conflicts of interest.

Authors’ contributions

The authors contributed equally to this review.

Acknowledgements

We thank the scientists who have worked with us and molded our ideas and views on HIV prevention and the microbiome, including Quarraisha Abdool Karim, CAPRISA and Columbia University; Adam Burgener, Public Health Agency of Canada; Nichole Klatt, University of Washington‐Seattle; Carolyn Williamson, CAPRISA & University of Cape Town, Sharon Hillier, Magee Womens Research Institute, University of Pittsburgh and Jeanne Marrazzo, University of Alabama at Birmingham School of Medicine.

Funding

Some of the research mentioned in the review was conducted as part of the DST‐NRF Centre of Excellence in HIV Prevention, which is supported by the Department of Science and Technology and the National Research Foundation.

Karim, S. S. A. , Baxter, C. , Passmore, J.‐A. S. , McKinnon, L. R. , Williams, B. L. The genital tract and rectal microbiomes: their role in HIV susceptibility and prevention in women. J Int AIDS Soc. 2019; 22(5):e25300

References

  • 1. UNAIDS . Miles to go: closing gaps, breaking barriers, righting injustices. Geneva, Switzerland: Joint United Nations Programme on HIV/AIDS (UNAIDS); 2018. [Google Scholar]
  • 2. Kharsany ABM, Cawood C, Khanyile D, Lewis L, Grobler A, Puren A, et al. Community‐based HIV prevalence in KwaZulu‐Natal, South Africa: results of a cross‐sectional household survey. Lancet HIV. 2018. 5(8):e427–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Abdool Karim Q, Sibeko S, Baxter C. Preventing HIV infection in women: a global health imperative. Clin Infect Dis. 2010;50 Suppl 3:S122–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164(3):337–40. [DOI] [PubMed] [Google Scholar]
  • 5. Halfvarson J, Brislawn CJ, Lamendella R, Vazquez‐Baeza Y, Walters WA, Bramer LM, et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat Microbiol. 2017;2:17004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Knights D, Parfrey LW, Zaneveld J, Lozupone C, Knight R. Human‐associated microbial signatures: examining their predictive value. Cell Host Microbe. 2011;10(4):292–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med. 2016;375(24):2369–79. [DOI] [PubMed] [Google Scholar]
  • 8. Gilbert JA, Blaser MJ, Caporaso JG, Jansson JK, Lynch SV, Knight R. Current understanding of the human microbiome. Nat Med. 2018;24(4):392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Hickey RJ, Zhou X, Pierson JD, Ravel J, Forney LJ. Understanding vaginal microbiome complexity from an ecological perspective. Transl Res. 2012;160(4):267–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Cheng J, et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science. 2013;339(6119):548–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Leffler DA, Lamont JT. Clostridium difficile infection. N Engl J Med. 2015;372(16):1539–48. [DOI] [PubMed] [Google Scholar]
  • 13. Smith WL, Hedges SR, Mordechai E, Adelson ME, Trama JP, Gygax SE, et al. Cervical and vaginal flora specimens are highly concordant with respect to bacterial vaginosis‐associated organisms and commensal Lactobacillus species in women of reproductive age. J Clin Microbiol. 2014;52(8):3078–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Chehoud C, Stieh DJ, Bailey AG, Laughlin AL, Allen SA, McCotter KL, et al. Associations of the vaginal microbiota with HIV infection, bacterial vaginosis, and demographic factors. AIDS. 2017;31(7):895–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Garcia‐Velasco JA, Menabrito M, Catalan IB. What fertility specialists should know about the vaginal microbiome: a review. Reprod Biomed Online. 2017;35(1):103–12. [DOI] [PubMed] [Google Scholar]
  • 16. Gajer P, Brotman RM, Bai G, Sakamoto J, Schutte UM, Zhong X, et al. Temporal dynamics of the human vaginal microbiota. Sci Transl Med. 2012;4(132):132ra52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Muhleisen AL, Herbst‐Kralovetz MM. Menopause and the vaginal microbiome. Maturitas. 2016;91:42–50. [DOI] [PubMed] [Google Scholar]
  • 18. Yamamoto T, Zhou X, Williams CJ, Hochwalt A, Forney LJ. Bacterial populations in the vaginas of healthy adolescent women. J Pediatr Adolesc Gynecol. 2009;22(1):11–8. [DOI] [PubMed] [Google Scholar]
  • 19. Eschenbach DA, Thwin SS, Patton DL, Hooton TM, Stapleton AE, Agnew K, et al. Influence of the normal menstrual cycle on vaginal tissue, discharge, and microflora. Clin Infect Dis. 2000;30(6):901–7. [DOI] [PubMed] [Google Scholar]
  • 20. Noyes N, Cho KC, Ravel J, Forney LJ, Abdo Z. Associations between sexual habits, menstrual hygiene practices, demographics and the vaginal microbiome as revealed by Bayesian network analysis. PLoS ONE. 2018;13(1):e0191625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Fethers KA, Fairley CK, Hocking JS, Gurrin LC, Bradshaw CS. Sexual risk factors and bacterial vaginosis: a systematic review and meta‐analysis. Clin Infect Dis. 2008;47(11):1426–35. [DOI] [PubMed] [Google Scholar]
  • 22. McClelland RS, Richardson BA, Graham SM, Masese LN, Gitau R, Lavreys L, et al. A prospective study of risk factors for bacterial vaginosis in HIV‐1‐seronegative African women. Sex Transm Dis. 2008;35(6):617–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Schwebke JR, Richey CM, Weiss HL. Correlation of behaviors with microbiological changes in vaginal flora. J Infect Dis. 1999;180(5):1632–6. [DOI] [PubMed] [Google Scholar]
  • 24. Brooks JP, Edwards DJ, Blithe DL, Fettweis JM, Serrano MG, Sheth NU, et al. Effects of combined oral contraceptives, depot medroxyprogesterone acetate and the levonorgestrel‐releasing intrauterine system on the vaginal microbiome. Contraception. 2017;95(4):405–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Albert AY, Chaban B, Wagner EC, Schellenberg JJ, Links MG, van Schalkwyk J, et al. A study of the vaginal microbiome in healthy Canadian women utilizing cpn60‐based molecular profiling reveals distinct Gardnerella subgroup community state types. PLoS ONE. 2015;10(8):e0135620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Brotman RM. Vaginal microbiome and sexually transmitted infections: an epidemiologic perspective. J Clin Invest. 2011;121(12):4610–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Stoyancheva G, Marzotto M, Dellaglio F, Torriani S. Bacteriocin production and gene sequencing analysis from vaginal Lactobacillus strains. Arch Microbiol. 2014;196(9):645–53. [DOI] [PubMed] [Google Scholar]
  • 28. Lai SK, Hida K, Shukair S, Wang YY, Figueiredo A, Cone R, et al. Human immunodeficiency virus type 1 is trapped by acidic but not by neutralized human cervicovaginal mucus. J Virol. 2009;83(21):11196–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. 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]
  • 30. Verstraelen H, Verhelst R, Claeys G, De Backer E, Temmerman M, Vaneechoutte M. 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] [PMC free article] [PubMed] [Google Scholar]
  • 31. Huang B, Fettweis JM, Brooks JP, Jefferson KK, Buck GA. The changing landscape of the vaginal microbiome. Clin Lab Med. 2014;34(4):747–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Ness RB, Hillier S, Richter HE, Soper DE, Stamm C, Bass DC, et al. Can known risk factors explain racial differences in the occurrence of bacterial vaginosis? J Natl Med Assoc. 2003;95(3):201–12. [PMC free article] [PubMed] [Google Scholar]
  • 33. Zhou X, Hansmann MA, Davis CC, Suzuki H, Brown CJ, Schutte U, et al. The vaginal bacterial communities of Japanese women resemble those of women in other racial groups. FEMS Immunol Med Microbiol. 2010;58(2):169–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Zhou X, Brown CJ, Abdo Z, Davis CC, Hansmann MA, Joyce P, et al. Differences in the composition of vaginal microbial communities found in healthy Caucasian and black women. ISME J. 2007;1(2):121–33. [DOI] [PubMed] [Google Scholar]
  • 35. Zhou X, Bent SJ, Schneider MG, Davis CC, Islam MR, Forney LJ. Characterization of vaginal microbial communities in adult healthy women using cultivation‐independent methods. Microbiology. 2004;150(Pt 8):2565–73. [DOI] [PubMed] [Google Scholar]
  • 36. Reid G. Has knowledge of the vaginal microbiome altered approaches to health and disease? F1000Res. 2018;7:460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Fettweis JM, Brooks JP, Serrano MG, Sheth NU, Girerd PH, Edwards DJ, et al. Differences in vaginal microbiome in African American women versus women of European ancestry. Microbiology. 2014;160(Pt 10):2272–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, et al. Vaginal microbiome of reproductive‐age women. Proc Natl Acad Sci USA. 2011;108 Suppl 1:4680–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Ravel J, Brotman RM. Translating the vaginal microbiome: gaps and challenges. Genome Med. 2016;8(1):35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Onderdonk AB, Delaney ML, Fichorova RN. The Human Microbiome during Bacterial Vaginosis. Clin Microbiol Rev. 2016;29(2):223–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Nasioudis D, Linhares IM, Ledger WJ, Witkin SS. Bacterial vaginosis: a critical analysis of current knowledge. BJOG. 2017;124(1):61–9. [DOI] [PubMed] [Google Scholar]
  • 42. Rodriguez Jovita M, Collins MD, Sjoden B, Falsen E. Characterization of a novel Atopobium isolate from the human vagina: description of Atopobium vaginae sp. nov. Int J Syst Bacteriol. 1999;49 Pt 4:1573–6. [DOI] [PubMed] [Google Scholar]
  • 43. Ma B, Forney LJ, Ravel J. Vaginal microbiome: rethinking health and disease. Annu Rev Microbiol. 2012;66:371–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Romero R, Hassan SS, Gajer P, Tarca AL, Fadrosh DW, Nikita L, et al. The composition and stability of the vaginal microbiota of normal pregnant women is different from that of non‐pregnant women. Microbiome. 2014;2(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Anahtar MN, Byrne EH, Doherty KE, Bowman BA, Yamamoto HS, Soumillon M, et al. Cervicovaginal bacteria are a major modulator of host inflammatory responses in the female genital tract. Immunity. 2015;42(5):965–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Balle C, Lennard K, Dabee S, Barnabas SL, Jaumdally SZ, Gasper MA, et al. Endocervical and vaginal microbiota in South African adolescents with asymptomatic Chlamydia trachomatis infection. Sci Rep. 2018;8(1):11109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Exley RM, Wu H, Shaw J, Schneider MC, Smith H, Jerse AE, et al. Lactate acquisition promotes successful colonization of the murine genital tract by Neisseria gonorrhoeae. Infect Immun. 2007;75(3):1318–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. 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]
  • 49. Lennard K, Dabee S, Barnabas SL, Havyarimana E, Blakney A, Jaumdally SZ, et al. Microbial composition predicts genital tract inflammation and persistent bacterial vaginosis in South African adolescent females. Infect Immun. 2018;86(1):e00410–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Brooks JP, Buck GA, Chen G, Diao L, Edwards DJ, Fettweis JM, et al. Changes in vaginal community state types reflect major shifts in the microbiome. Microb Ecol Health Dis. 2017;28(1):1303265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Verhelst R, Verstraelen H, Claeys G, Verschraegen G, Van Simaey L, De Ganck C, et al. Comparison between Gram stain and culture for the characterization of vaginal microflora: definition of a distinct grade that resembles grade I microflora and revised categorization of grade I microflora. BMC Microbiol. 2005;5:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Human Microbiome Project Consortium . Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Hummelen R, Fernandes AD, Macklaim JM, Dickson RJ, Changalucha J, Gloor GB, et al. Deep sequencing of the vaginal microbiota of women with HIV. PLoS ONE. 2010;5(8):e12078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature. 2011;473(7346):174–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Witkin SS, Ledger WJ. Complexities of the uniquely human vagina. Sci Transl Med. 2012;4(132):132fs11. [DOI] [PubMed] [Google Scholar]
  • 56. Eschenbach DA, Davick PR, Williams BL, Klebanoff SJ, Young‐Smith K, Critchlow CM, et al. Prevalence of hydrogen peroxide‐producing Lactobacillus species in normal women and women with bacterial vaginosis. J Clin Microbiol. 1989;27(2):251–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Srinivasan S, Hoffman NG, Morgan MT, Matsen FA, Fiedler TL, Hall RW, et al. Bacterial communities in women with bacterial vaginosis: high resolution phylogenetic analyses reveal relationships of microbiota to clinical criteria. PLoS ONE. 2012;7(6):e37818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. 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(1):12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Eschenbach DA. History and review of bacterial vaginosis. Am J Obstet Gynecol. 1993;169 2 Pt 2:441–5. [DOI] [PubMed] [Google Scholar]
  • 60. Taha TE, Hoover DR, Dallabetta GA, Kumwenda NI, Mtimavalye LA, Yang LP, et al. Bacterial vaginosis and disturbances of vaginal flora: association with increased acquisition of HIV. AIDS. 1998;12(13):1699–706. [DOI] [PubMed] [Google Scholar]
  • 61. Marrazzo JM. Interpreting the epidemiology and natural history of bacterial vaginosis: are we still confused? Anaerobe. 2011;17(4):186–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. 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(1):14–22. [DOI] [PubMed] [Google Scholar]
  • 63. Klebanoff MA, Schwebke JR, Zhang J, Nansel TR, Yu KF, Andrews WW. Vulvovaginal symptoms in women with bacterial vaginosis. Obstet Gynecol. 2004;104(2):267–72. [DOI] [PubMed] [Google Scholar]
  • 64. Nugent RP, Krohn MA, Hillier SL. Reliability of diagnosing bacterial vaginosis is improved by a standardized method of gram stain interpretation. J Clin Microbiol. 1991;29(2):297–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Kenyon CR, Osbak K. Recent progress in understanding the epidemiology of bacterial vaginosis. Curr Opin Obstet Gynecol. 2014;26(6):448–54. [DOI] [PubMed] [Google Scholar]
  • 66. Yeoman CJ, Yildirim S, Thomas SM, Durkin AS, Torralba M, Sutton G, et al. Comparative genomics of Gardnerella vaginalis strains reveals substantial differences in metabolic and virulence potential. PLoS ONE. 2010;5(8):e12411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Criswell BS, Ladwig CL, Gardner HL, Dukes CD. Haemophilus vaginalis: vaginitis by inoculation from culture. Obstet Gynecol. 1969;33:195–9. [PubMed] [Google Scholar]
  • 68. Smart S, Singal A, Mindel A. Social and sexual risk factors for bacterial vaginosis. Sex Transm Infect. 2004;80(1):58–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Gallo MF, Macaluso M, Warner L, Fleenor ME, Hook EW III, Brill I, et al. Bacterial vaginosis, gonorrhea, and chlamydial infection among women attending a sexually transmitted disease clinic: a longitudinal analysis of possible causal links. Ann Epidemiol. 2012;22(3):213–20. [DOI] [PubMed] [Google Scholar]
  • 70. Brotman RM, Ravel J, Cone RA, Zenilman JM. Rapid fluctuation of the vaginal microbiota measured by Gram stain analysis. Sex Transm Infect. 2010;86(4):297–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Cherpes TL, Hillier SL, Meyn LA, Busch JL, Krohn MA. A delicate balance: risk factors for acquisition of bacterial vaginosis include sexual activity, absence of hydrogen peroxide‐producing lactobacilli, black race, and positive herpes simplex virus type 2 serology. Sex Transm Dis. 2008;35(1):78–83. [DOI] [PubMed] [Google Scholar]
  • 72. Zozaya M, Ferris MJ, Siren JD, Lillis R, Myers L, Nsuami MJ, 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]
  • 73. Baud D, Pattaroni C, Vulliemoz N, Castella V, Marsland BJ, Stojanov M. sperm microbiota and its impact on semen parameters. Front Microbiol. 2019;10:234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Amaya‐Guio J, Viveros‐Carreno DA, Sierra‐Barrios EM, Martinez‐Velasquez MY, Grillo‐Ardila CF. Antibiotic treatment for the sexual partners of women with bacterial vaginosis. Cochrane Database Syst Rev. 2016;10:CD011701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Plummer EL, Vodstrcil LA, Danielewski JA, Murray GL, Fairley CK, Garland SM, et al. Combined oral and topical antimicrobial therapy for male partners of women with bacterial vaginosis: acceptability, tolerability and impact on the genital microbiota of couples – a pilot study. PLoS ONE. 2018;13(1):e0190199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Turovskiy Y, Sutyak Noll K, Chikindas ML. The aetiology of bacterial vaginosis. J Appl Microbiol. 2011;110(5):1105–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Tao L, Pavlova SI, Mou SM, Ma WG, Kilic AO. Analysis of lactobacillus products for phages and bacteriocins that inhibit vaginal lactobacilli. Infect Dis Obstet Gynecol. 1997;5(3):244–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Blackwell AL. Vaginal bacterial phaginosis? Sex Transm Infect. 1999;75(5):352–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Kilic AO, Pavlova SI, Ma WG, Tao L. Analysis of Lactobacillus phages and bacteriocins in American dairy products and characterization of a phage isolated from yogurt. Appl Environ Microbiol. 1996;62(6):2111–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Raya RR, Kleeman EG, Luchansky JB, Klaenhammer TR. Characterization of the temperate bacteriophage phi adh and plasmid transduction in Lactobacillus acidophilus ADH. Appl Environ Microbiol. 1989;55(9):2206–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Pavlova SI, Kilic AO, Mou SM, Tao L. Phage infection in vaginal lactobacilli: an in vitro study. Infect Dis Obstet Gynecol. 1997;5(1):36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Blatny JM, Godager L, Lunde M, Nes IF. Complete genome sequence of the Lactococcus lactis temperate phage phiLC3: comparative analysis of phiLC3 and its relatives in lactococci and streptococci. Virology. 2004;318(1):231–44. [DOI] [PubMed] [Google Scholar]
  • 83. Masson L, Arnold KB, Little F, Mlisana K, Lewis DA, Mkhize N, et al. Inflammatory cytokine biomarkers to identify women with asymptomatic sexually transmitted infections and bacterial vaginosis who are at high risk of HIV infection. Sex Transm Infect. 2016;92(3):186–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Arnold KB, Burgener A, Birse K, Romas L, Dunphy LJ, Shahabi K, et al. Increased levels of inflammatory cytokines in the female reproductive tract are associated with altered expression of proteases, mucosal barrier proteins, and an influx of HIV‐susceptible target cells. Mucosal Immunol. 2016;9(1):194–205. [DOI] [PubMed] [Google Scholar]
  • 85. McKinnon LR, Liebenberg LJ, Yende‐Zuma N, Archary D, Ngcapu S, Sivro A, et al. Genital inflammation undermines the effectiveness of tenofovir gel in preventing HIV acquisition in women. Nat Med. 2018;24(4):491–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Li Q, Estes JD, Schlievert PM, Duan L, Brosnahan AJ, Southern PJ, et al. Glycerol monolaurate prevents mucosal SIV transmission. Nature. 2009;458(7241):1034–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Osborn L, Kunkel S, Nabel GJ. Tumor necrosis factor alpha and interleukin 1 stimulate the human immunodeficiency virus enhancer by activation of the nuclear factor kappa B. Proc Natl Acad Sci USA. 1989;86(7):2336–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Nazli A, Chan O, Dobson‐Belaire WN, Ouellet M, Tremblay MJ, Gray‐Owen SD, et al. Exposure to HIV‐1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Pathog. 2010;6(4):e1000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Zevin AS, Xie IY, Birse K, Arnold K, Romas L, Westmacott G, et al. Microbiome composition and function drives wound‐healing impairment in the female genital tract. PLoS Pathog. 2016;12(9):e1005889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Klatt NR, Cheu R, Birse K, Zevin AS, Perner M, Noel‐Romas L, et al. Vaginal bacteria modify HIV tenofovir microbicide efficacy in African women. Science. 2017;356(6341):938–45. [DOI] [PubMed] [Google Scholar]
  • 91. Williams B, Passmore J‐A. Role of vaginal microbiota in genital inflammation and enhancing HIV acquisition in women. AIDS 2016; 2016 18‐22 July; Durban, South Africa; 2016.
  • 92. Gelber SE, Aguilar JL, Lewis KL, Ratner AJ. Functional and phylogenetic characterization of Vaginolysin, the human‐specific cytolysin from Gardnerella vaginalis. J Bacteriol. 2008;190(11):3896–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Cauci S, Culhane JF, Di Santolo M, McCollum K. Among pregnant women with bacterial vaginosis, the hydrolytic enzymes sialidase and prolidase are positively associated with interleukin‐1beta. Am J Obstet Gynecol. 2008;198(1):132.e1–7. [DOI] [PubMed] [Google Scholar]
  • 94. Africa CW, Nel J, Stemmet M. Anaerobes and bacterial vaginosis in pregnancy: virulence factors contributing to vaginal colonisation. Int J Environ Res Public Health. 2014;11(7):6979–7000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Nasu K, Narahara H. Pattern recognition via the toll‐like receptor system in the human female genital tract. Mediators Inflamm. 2010;2010:976024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Pioli PA, Amiel E, Schaefer TM, Connolly JE, Wira CR, Guyre PM. Differential expression of Toll‐like receptors 2 and 4 in tissues of the human female reproductive tract. Infect Immun. 2004;72(10):5799–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Lin Z, Xu J, Jin X, Zhang X, Ge F. Modulation of expression of Toll‐like receptors in the human endometrium. Am J Reprod Immunol. 2009;61(5):338–45. [DOI] [PubMed] [Google Scholar]
  • 98. Aroutcheva A, Ling Z, Faro S. Prevotella bivia as a source of lipopolysaccharide in the vagina. Anaerobe. 2008;14(5):256–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Kim SJ, Ha MS, Choi EY, Choi JI, Choi IS. Nitric oxide production and inducible nitric oxide synthase expression induced by Prevotella nigrescens lipopolysaccharide. FEMS Immunol Med Microbiol. 2005;43(1):51–8. [DOI] [PubMed] [Google Scholar]
  • 100. Hill GB. The microbiology of bacterial vaginosis. Am J Obstet Gynecol. 1993;169 2 Pt 2:450–4. [DOI] [PubMed] [Google Scholar]
  • 101. Hay PE, Lamont RF, Taylor‐Robinson D, Morgan DJ, Ison C, Pearson J. Abnormal bacterial colonisation of the genital tract and subsequent preterm delivery and late miscarriage. BMJ. 1994;308(6924):295–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Tecce MG, Basta MN, Shubinets V, Lanni MA, Mirzabeigi MN, Cooney L, et al. A risk model and cost analysis of post‐operative incisional hernia following 2,145 open hysterectomies‐Defining indications and opportunities for risk reduction. Am J Surg. 2017;213(6):1083–90. [DOI] [PubMed] [Google Scholar]
  • 103. van de Wijgert JH, Morrison CS, Cornelisse PG, Munjoma M, Moncada J, Awio P, et al. Bacterial vaginosis and vaginal yeast, but not vaginal cleansing, increase HIV‐1 acquisition in African women. J Acquir Immune Defic Syndr. 2008;48(2):203–10. [DOI] [PubMed] [Google Scholar]
  • 104. Nelson DB, Hanlon A, Nachamkin I, Haggerty C, Mastrogiannis DS, Liu C, et al. Early pregnancy changes in bacterial vaginosis‐associated bacteria and preterm delivery. Paediatr Perinat Epidemiol. 2014;28(2):88–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Cohen CR, Duerr A, Pruithithada N, Rugpao S, Hillier S, Garcia P, et al. Bacterial vaginosis and HIV seroprevalence among female commercial sex workers in Chiang Mai, Thailand. AIDS. 1995;9(9):1093–7. [DOI] [PubMed] [Google Scholar]
  • 106. Coleman JS, Hitti J, Bukusi EA, Mwachari C, Muliro A, Nguti R, et al. Infectious correlates of HIV‐1 shedding in the female upper and lower genital tracts. AIDS. 2007;21(6):755–9. [DOI] [PubMed] [Google Scholar]
  • 107. Cu‐Uvin S, Hogan JW, Caliendo AM, Harwell J, Mayer KH, Carpenter CC, et al. Association between bacterial vaginosis and expression of human immunodeficiency virus type 1 RNA in the female genital tract. Clin Infect Dis. 2001;33(6):894–6. [DOI] [PubMed] [Google Scholar]
  • 108. Martin HL, Richardson BA, Nyange PM, Lavreys L, Hillier SL, Chohan B, et al. Vaginal lactobacilli, microbial flora, and risk of human immunodeficiency virus type 1 and sexually transmitted disease acquisition. J Infect Dis. 1999;180(6):1863–8. [DOI] [PubMed] [Google Scholar]
  • 109. Spear GT, St John E, Zariffard MR. Bacterial vaginosis and human immunodeficiency virus infection. AIDS Res Ther. 2007;4:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Borgdorff H, Tsivtsivadze E, Verhelst R, Marzorati M, Jurriaans S, Ndayisaba GF, et al. Lactobacillus‐dominated cervicovaginal microbiota associated with reduced HIV/STI prevalence and genital HIV viral load in African women. ISME J. 2014;8(9):1781–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Gosmann C, Anahtar MN, Handley SA, Farcasanu M, Abu‐Ali G, Bowman BA, et al. Lactobacillus‐deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African women. Immunity. 2017;46(1):29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Bradshaw CS, Morton AN, Hocking J, Garland SM, Morris MB, Moss LM, 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(11):1478–86. [DOI] [PubMed] [Google Scholar]
  • 113. Carmody RN, Turnbaugh PJ. Host‐microbial interactions in the metabolism of therapeutic and diet‐derived xenobiotics. J Clin Invest. 2014;124(10):4173–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Koppel N, Maini Rekdal V, Balskus EP. Chemical transformation of xenobiotics by the human gut microbiota. Science. 2017;356(6344):eaag2770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Sousa T, Paterson R, Moore V, Carlsson A, Abrahamsson B, Basit AW. The gastrointestinal microbiota as a site for the biotransformation of drugs. Int J Pharm. 2008;363(1–2):1–25. [DOI] [PubMed] [Google Scholar]
  • 116. Danielsson H, Gustafsson B. On serum‐cholesterol levels and neutral fecal sterols in germ‐free rats; bile acids and steroids 59. Arch Biochem Biophys. 1959;83:482–5. [DOI] [PubMed] [Google Scholar]
  • 117. Ravel J, Brotman RM, Gajer P, Ma B, Nandy M, Fadrosh DW, et al. Daily temporal dynamics of vaginal microbiota before, during and after episodes of bacterial vaginosis. Microbiome. 2013;1(1):29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Eschenbach DA, Patton DL, Meier A, Thwin SS, Aura J, Stapleton A, et al. Effects of oral contraceptive pill use on vaginal flora and vaginal epithelium. Contraception. 2000;62(3):107–12. [DOI] [PubMed] [Google Scholar]
  • 119. Braundmeier AG, Lenz KM, Inman KS, Chia N, Jeraldo P, Walther‐Antonio MR, et al. Individualized medicine and the microbiome in reproductive tract. Front Physiol. 2015;6:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Srinivasan S, Liu C, Mitchell CM, Fiedler TL, Thomas KK, Agnew KJ, et al. Temporal variability of human vaginal bacteria and relationship with bacterial vaginosis. PLoS ONE. 2010;5(4):e10197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Vodstrcil LA, Hocking JS, Law M, Walker S, Tabrizi SN, Fairley CK, et al. Hormonal contraception is associated with a reduced risk of bacterial vaginosis: a systematic review and meta‐analysis. PLoS ONE. 2013;8(9):e73055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Bautista CT, Wurapa E, Sateren WB, Morris S, Hollingsworth B, Sanchez JL. Bacterial vaginosis: a synthesis of the literature on etiology, prevalence, risk factors, and relationship with chlamydia and gonorrhea infections. Mil Med Res. 2016;3:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. DiGiulio DB, Callahan BJ, McMurdie PJ, Costello EK, Lyell DJ, Robaczewska A, et al. Temporal and spatial variation of the human microbiota during pregnancy. Proc Natl Acad Sci USA. 2015;112(35):11060–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Sjoberg I, Cajander S, Rylander E. Morphometric characteristics of the vaginal epithelium during the menstrual cycle. Gynecol Obstet Invest. 1988;26(2):136–44. [DOI] [PubMed] [Google Scholar]
  • 125. Sanchez S, Garcia PJ, Thomas KK, Catlin M, Holmes KK. Intravaginal metronidazole gel versus metronidazole plus nystatin ovules for bacterial vaginosis: a randomized controlled trial. Am J Obstet Gynecol. 2004;191(6):1898–906. [DOI] [PubMed] [Google Scholar]
  • 126. Schwebke JR, Desmond R. Risk factors for bacterial vaginosis in women at high risk for sexually transmitted diseases. Sex Transm Dis. 2005;32(11):654–8. [DOI] [PubMed] [Google Scholar]
  • 127. Gray RH, Kigozi G, Serwadda D, Makumbi F, Nalugoda F, Watya S, 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(1):42.e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Huang H, Song L, Zhao W. Effects of probiotics for the treatment of bacterial vaginosis in adult women: a meta‐analysis of randomized clinical trials. Arch Gynecol Obstet. 2014;289(6):1225–34. [DOI] [PubMed] [Google Scholar]
  • 129. Tan H, Fu Y, Yang C, Ma J. Effects of metronidazole combined probiotics over metronidazole alone for the treatment of bacterial vaginosis: a meta‐analysis of randomized clinical trials. Arch Gynecol Obstet. 2017;295(6):1331–9. [DOI] [PubMed] [Google Scholar]
  • 130. Happel AU, Jaumdally SZ, Pidwell T, Cornelius T, Jaspan HB, Froissart R, et al. Probiotics for vaginal health in South Africa: what is on retailers’ shelves? BMC Womens Health. 2017;17(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Hemmerling A, Cohen CR. Probiotics: the potential for a live microbicide to prevent HIV. J Acquir Immune Defic Syndr. 2011;56(3):e98–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Hemmerling A, Harrison W, Schroeder A, Park J, Korn A, Shiboski S, et al. Phase 2a study assessing colonization efficiency, safety, and acceptability of Lactobacillus crispatus CTV‐05 in women with bacterial vaginosis. Sex Transm Dis. 2010;37(12):745–50. [DOI] [PubMed] [Google Scholar]
  • 133. Saleh M, Elson CO. Experimental inflammatory bowel disease: insights into the host‐microbiota dialog. Immunity. 2011;34(3):293–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157(1):121–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200(6):749–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Mehandru S, Poles MA, Tenner‐Racz K, Horowitz A, Hurley A, Hogan C, et al. Primary HIV‐1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200(6):761–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71. [DOI] [PubMed] [Google Scholar]
  • 139. Ellis CL, Ma ZM, Mann SK, Li CS, Wu J, Knight TH, et al. Molecular characterization of stool microbiota in HIV‐infected subjects by panbacterial and order‐level 16S ribosomal DNA (rDNA) quantification and correlations with immune activation. J Acquir Immune Defic Syndr. 2011;57(5):363–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Mutlu EA, Keshavarzian A, Losurdo J, Swanson G, Siewe B, Forsyth C, et al. A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjects. PLoS Pathog. 2014;10(2):e1003829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Perez‐Santiago J, Gianella S, Massanella M, Spina CA, Karris MY, Var SR, et al. Gut Lactobacillales are associated with higher CD4 and less microbial translocation during HIV infection. AIDS. 2013;27(12):1921–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Dillon SM, Lee EJ, Kotter CV, Austin GL, Gianella S, Siewe B, et al. Gut dendritic cell activation links an altered colonic microbiome to mucosal and systemic T‐cell activation in untreated HIV‐1 infection. Mucosal Immunol. 2016;9(1):24–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Vazquez‐Castellanos JF, Serrano‐Villar S, Latorre A, Artacho A, Ferrus ML, Madrid N, et al. Altered metabolism of gut microbiota contributes to chronic immune activation in HIV‐infected individuals. Mucosal Immunol. 2015;8(4):760–72. [DOI] [PubMed] [Google Scholar]
  • 144. Serrano‐Villar S, Rojo D, Martinez‐Martinez M, Deusch S, Vazquez‐Castellanos JF, Bargiela R, et al. Gut bacteria metabolism impacts immune recovery in HIV‐infected individuals. EBioMedicine. 2016;8:203–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Noguera‐Julian M, Rocafort M, Guillen Y, Rivera J, Casadella M, Nowak P, et al. Gut microbiota linked to sexual preference and HIV infection. EBioMedicine. 2016;5:135–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Kelley CF, Kraft CS, de Man TJ, Duphare C, Lee HW, Yang J, et al. The rectal mucosa and condomless receptive anal intercourse in HIV‐negative MSM: implications for HIV transmission and prevention. Mucosal Immunol. 2017;10(4):996–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Kurilshikov A, Wijmenga C, Fu J, Zhernakova A. Host genetics and Gut microbiome: challenges and perspectives. Trends Immunol. 2017;38(9):633–47. [DOI] [PubMed] [Google Scholar]
  • 148. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, et al. Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell. 2015;163(2):367–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Stieh DJ, Matias E, Xu H, Fought AJ, Blanchard JL, Marx PA, et al. Th17 cells are preferentially infected very early after vaginal transmission of SIV in macaques. Cell Host Microbe. 2016;19(4):529–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. McKinnon LR, Nyanga B, Kim CJ, Izulla P, Kwatampora J, Kimani M, et al. Early HIV‐1 infection is associated with reduced frequencies of cervical Th17 cells. J Acquir Immune Defic Syndr. 2015;68(1):6–12. [DOI] [PubMed] [Google Scholar]
  • 151. Sui Y, Dzutsev A, Venzon D, Frey B, Thovarai V, Trinchieri G, et al. Influence of gut microbiome on mucosal immune activation and SHIV viral transmission in naive macaques. Mucosal Immunol. 2018;11(4):1219–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Hillier SL, Meyn LA, Bunge K, Austin M, Moncla BJ, Dezzutti CS, et al. Impact of vaginal microbiota on genital tissue and plasma concentrations of tenofovir. Conference of Retroviruses and Opportunistic Infections (CROI), February 13‐16. Seattle, Washington; 2017.
  • 153. Klatt NR. The vaginal microbiome and acquisition of HIV infection. Conference on Retroviruses and Opportunistic Infections [abstract 64]; March 4–7, 2018; Boston, Massachusetts; 2018.
  • 154. Taneva E, Sinclair S, Mesquita PM, Weinrick B, Cameron SA, Cheshenko N, et al. Vaginal microbiome modulates topical antiretroviral drug pharmacokinetics. JCI Insight. 2018;3(13):99545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Heffron R, McClelland RS, Balkus JE, Celum C, Cohen CR, Mugo N, et al. Efficacy of oral pre‐exposure prophylaxis (PrEP) for HIV among women with abnormal vaginal microbiota: a post‐hoc analysis of the randomised, placebo‐controlled Partners PrEP Study. Lancet HIV. 2017;4(10):e449–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Fredricks DN. Molecular methods to describe the spectrum and dynamics of the vaginal microbiota. Anaerobe. 2011;17(4):191–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Schellenberg J, Links MG, Hill JE, Dumonceaux TJ, Peters GA, Tyler S, et al. Pyrosequencing of the chaperonin‐60 universal target as a tool for determining microbial community composition. Appl Environ Microbiol. 2009;75(9):2889–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Hill JE, Goh SH, Money DM, Doyle M, Li A, Crosby WL, et al. Characterization of vaginal microflora of healthy, nonpregnant women by chaperonin‐60 sequence‐based methods. Am J Obstet Gynecol. 2005;193 3 Pt 1:682–92. [DOI] [PubMed] [Google Scholar]
  • 159. Fredricks DN, Fiedler TL, Marrazzo JM. Molecular identification of bacteria associated with bacterial vaginosis. N Engl J Med. 2005;353(18):1899–911. [DOI] [PubMed] [Google Scholar]
  • 160. Oakley BB, Fiedler TL, Marrazzo JM, Fredricks DN. Diversity of human vaginal bacterial communities and associations with clinically defined bacterial vaginosis. Appl Environ Microbiol. 2008;74(15):4898–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Austin MN, Rabe LK, Srinivasan S, Fredricks DN, Wiesenfeld HC, Hillier SL. Mageeibacillus indolicus gen. nov., sp. nov.: a novel bacterium isolated from the female genital tract. Anaerobe. 2015;32:37–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Paramel Jayaprakash T, Schellenberg JJ, Hill JE. Resolution and characterization of distinct cpn60‐based subgroups of Gardnerella vaginalis in the vaginal microbiota. PLoS ONE. 2012;7(8):e43009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Schellenberg JJ, Paramel Jayaprakash T, Withana Gamage N, Patterson MH, Vaneechoutte M, Hill JE. Gardnerella vaginalis Subgroups Defined by cpn60 Sequencing and Sialidase Activity in Isolates from Canada, Belgium and Kenya. PLoS ONE. 2016;11(1):e0146510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Freitas AC, Bocking A, Hill JE, Money DM; Group VR . Increased richness and diversity of the vaginal microbiota and spontaneous preterm birth. Microbiome. 2018; 6(1):117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Janulaitiene M, Paliulyte V, Grinceviciene S, Zakareviciene J, Vladisauskiene A, Marcinkute A, et al. Prevalence and distribution of Gardnerella vaginalis subgroups in women with and without bacterial vaginosis. BMC Infect Dis. 2017;17(1):394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Janulaitiene M, Gegzna V, Baranauskiene L, Bulavaite A, Simanavicius M, Pleckaityte M. Phenotypic characterization of Gardnerella vaginalis subgroups suggests differences in their virulence potential. PLoS ONE. 2018;13(7):e0200625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. McClelland RS, Lingappa JR, Srinivasan S, Kinuthia J, John‐Stewart GC, Jaoko W, et al. Evaluation of the association between the concentrations of key vaginal bacteria and the increased risk of HIV acquisition in African women from five cohorts: a nested case‐control study. Lancet Infect Dis. 2018;18(5):554–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Goltsman DSA, Sun CL, Proctor DM, DiGiulio DB, Robaczewska A, Thomas BC, et al. Metagenomic analysis with strain‐level resolution reveals fine‐scale variation in the human pregnancy microbiome. Genome Res. 2018;28(10):1467–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Truong DT, Franzosa EA, Tickle TL, Scholz M, Weingart G, Pasolli E, et al. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods. 2015;12(10):902–3. [DOI] [PubMed] [Google Scholar]
  • 170. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 2014;15(3):R46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Sangwan N, Xia F, Gilbert JA. Recovering complete and draft population genomes from metagenome datasets. Microbiome. 2016;4:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Roachford OSE, Nelson KE, Mohapatra BR. Comparative genomics of four Mycoplasma species of the human urogenital tract: analysis of their core genomes and virulence genes. Int J Med Microbiol. 2017;307(8):508–20. [DOI] [PubMed] [Google Scholar]
  • 174. Abdelmaksoud AA, Koparde VN, Sheth NU, Serrano MG, Glascock AL, Fettweis JM, et al. Comparison of Lactobacillus crispatus isolates from Lactobacillus‐dominated vaginal microbiomes with isolates from microbiomes containing bacterial vaginosis‐associated bacteria. Microbiology. 2016;162(3):466–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Ahmed A, Earl J, Retchless A, Hillier SL, Rabe LK, Cherpes TL, et al. Comparative genomic analyses of 17 clinical isolates of Gardnerella vaginalis provide evidence of multiple genetically isolated clades consistent with subspeciation into genovars. J Bacteriol. 2012;194(15):3922–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Ojala T, Kankainen M, Castro J, Cerca N, Edelman S, Westerlund‐Wikstrom B, et al. Comparative genomics of Lactobacillus crispatus suggests novel mechanisms for the competitive exclusion of Gardnerella vaginalis. BMC Genom. 2014;15:1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Twin J, Bradshaw CS, Garland SM, Fairley CK, Fethers K, Tabrizi SN. The potential of metatranscriptomics for identifying screening targets for bacterial vaginosis. PLoS ONE. 2013;8(9):e76892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Deng ZL, Gottschick C, Bhuju S, Masur C, Abels C, Wagner‐Dobler I Metatranscriptome analysis of the vaginal microbiota reveals potential mechanisms for protection against metronidazole in bacterial vaginosis. mSphere. 2018;3(3):e00262–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Castro J, Franca A, Bradwell KR, Serrano MG, Jefferson KK, Cerca N. Comparative transcriptomic analysis of Gardnerella vaginalis biofilms vs. planktonic cultures using RNA‐seq. NPJ Biofilms Microbiomes. 2017;3:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Berard AR, Perner M, Mutch S, Farr Zuend C, McQueen P, Burgener AD. Understanding mucosal and microbial functionality of the female reproductive tract by metaproteomics: implications for HIV transmission. Am J Reprod Immunol. 2018;e12977. [DOI] [PubMed] [Google Scholar]
  • 181. Marin E, Haesaert A, Padilla L, Adan J, Hernaez ML, Monteoliva L, et al. Unraveling gardnerella vaginalis surface proteins using cell shaving proteomics. Front Microbiol. 2018;9:975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Tanca A, Abbondio M, Palomba A, Fraumene C, Manghina V, Cucca F, et al. Potential and active functions in the gut microbiota of a healthy human cohort. Microbiome. 2017;5(1):79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Heintz‐Buschart A, May P, Laczny CC, Lebrun LA, Bellora C, Krishna A, et al. Integrated multi‐omics of the human gut microbiome in a case study of familial type 1 diabetes. Nat Microbiol. 2016;2:16180. [DOI] [PubMed] [Google Scholar]
  • 184. Pruski P, Lewis HV, Lee YS, Marchesi JR, Bennett PR, Takats Z, et al. Assessment of microbiota: host interactions at the vaginal mucosa interface. Methods. 2018;149:74–84. [DOI] [PubMed] [Google Scholar]
  • 185. Watson E, Reid G. Metabolomics as a clinical testing method for the diagnosis of vaginal dysbiosis. Am J Reprod Immunol. 2018;80(2):e12979. [DOI] [PubMed] [Google Scholar]
  • 186. Yeoman CJ, Thomas SM, Miller ME, Ulanov AV, Torralba M, Lucas S, et al. A multi‐omic systems‐based approach reveals metabolic markers of bacterial vaginosis and insight into the disease. PLoS ONE. 2013;8(2):e56111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Srinivasan S, Morgan MT, Fiedler TL, Djukovic D, Hoffman NG, Raftery D, et al. Metabolic signatures of bacterial vaginosis. MBio. 2015;6(2):e00204–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Vitali B, Cruciani F, Picone G, Parolin C, Donders G, Laghi L. Vaginal microbiome and metabolome highlight specific signatures of bacterial vaginosis. Eur J Clin Microbiol Infect Dis. 2015;34(12):2367–76. [DOI] [PubMed] [Google Scholar]
  • 189. McMillan A, Rulisa S, Sumarah M, Macklaim JM, Renaud J, Bisanz JE, et al. A multi‐platform metabolomics approach identifies highly specific biomarkers of bacterial diversity in the vagina of pregnant and non‐pregnant women. Sci Rep. 2015;5:14174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Noecker C, Eng A, Srinivasan S, Theriot CM, Young VB, Jansson JK, et al. Metabolic model‐based integration of microbiome taxonomic and metabolomic profiles elucidates mechanistic links between ecological and metabolic variation. mSystems. 2016;1(1):e00013–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Parolin C, Foschi C, Laghi L, Zhu C, Banzola N, Gaspari V, et al. Insights into vaginal bacterial communities and metabolic profiles of chlamydia trachomatis infection: positioning between eubiosis and dysbiosis. Front Microbiol. 2018;9:600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Jordan SJ, Olson KM, Barnes S, Wilson LS, Berryhill TF, Bakshi R, et al. Lower levels of cervicovaginal tryptophan are associated with natural clearance of chlamydia in women. J Infect Dis. 2017;215(12):1888–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Goytia M, Shafer WM. Polyamines can increase resistance of Neisseria gonorrhoeae to mediators of the innate human host defense. Infect Immun. 2010;78(7):3187–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Laghi L, Picone G, Cruciani F, Brigidi P, Calanni F, Donders G, et al. Rifaximin modulates the vaginal microbiome and metabolome in women affected by bacterial vaginosis. Antimicrob Agents Chemother. 2014;58(6):3411–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Walsh J, Griffin BT, Clarke G, Hyland NP. Drug‐gut microbiota interactions: implications for neuropharmacology. Br J Pharmacol. 2018;175(24):4415–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Vazquez‐Baeza Y, Callewaert C, Debelius J, Hyde E, Marotz C, Morton JT, et al. Impacts of the human Gut microbiome on therapeutics. Annu Rev Pharmacol Toxicol. 2018;58:253–70. [DOI] [PubMed] [Google Scholar]
  • 197. Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65. [DOI] [PubMed] [Google Scholar]
  • 198. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, et al. Gut microbiome influences efficacy of PD‐1‐based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7. [DOI] [PubMed] [Google Scholar]
  • 199. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti‐PD‐1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti‐PD‐1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Brotman RM, Shardell MD, Gajer P, Tracy JK, Zenilman JM, Ravel J, et al. Interplay between the temporal dynamics of the vaginal microbiota and human papillomavirus detection. J Infect Dis. 2014;210(11):1723–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Camacho DM, Collins KM, Powers RK, Costello JC, Collins JJ. Next‐generation machine learning for biological networks. Cell. 2018;173(7):1581–92. [DOI] [PubMed] [Google Scholar]
  • 203. Lum PY, Singh G, Lehman A, Ishkanov T, Vejdemo‐Johansson M, Alagappan M, et al. Extracting insights from the shape of complex data using topology. Sci Rep. 2013;3:1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Zhang W, Chien J, Yong J, Kuang R. Network‐based machine learning and graph theory algorithms for precision oncology. NPJ Precis Oncol. 2017;1(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Huang S, Chaudhary K, Garmire LX. More is better: recent progress in multi‐omics data integration methods. Front Genet. 2017;8:84. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of the International AIDS Society are provided here courtesy of Wiley

RESOURCES