Skip to main content

Cervicovaginal microbiota: a promising direction for prevention and treatment in cervical cancer

Abstract

Cervical cancer is a common malignancy in women, with high incidence rate and mortality. Persistent infection of high-risk human papillomavirus (HPV) is the most important risk factor for cervical cancer and precancerous lesions. Cervicovaginal microbiota (CVM) plays an essential role in the defense of HPV infections and prevention of subsequent lesions. Dominance of Lactobacillus is the key of CVM homeostasis, which can be regulated by host, exogenous and endogenous factors. Dysbiosis of CVM, including altered microbial, metabolic, and immune signatures, can contribute to persist HPV infection, leading to cervical cancer. However, there is no evidence of the causality between CVM and cervical cancer, and the underlying mechanism remains unexplored. Considering the close correlation between CVM dysbiosis and persistent HPV infection, this review will overview CVM, its role in cervical cancer development and related mechanisms, and the prospects for therapeutic applications.

Introduction

Cervical cancer (CC) is the fourth most frequent cancer in women globally with estimated 604 000 new cases and 342 000 deaths in 2020 [1]. It is estimated to be approximately 553 000 new cases and 229 000 deaths worldwide in 2024 [2, 3]. Infection of high-risk human papilloma virus (HPV), mainly HPV-16 and HPV-18, is recognized as a significant carcinogenic factor of cervical cancer [4, 5]. Although 85 − 90% of high-risk HPV (hrHPV) infections can be spontaneously cleared within 6 months, a few HPV infections will still persist, leading to cervical intraepithelial neoplasia (CIN), also-called squamous intraepithelial lesion (SIL), and ultimately invasive cervical cancer [4, 6]. The fact suggests the presence of other factors involved in the development of cervical cancer.

Cervicovaginal microbiota (CVM), as a protective barrier for female reproductive system, plays an essential role in the defense against several primary and opportunistic pathogens including sexually transmitted infections (STIs) [7], especially HPVs [6]. A healthy CVM can form an acidic microenvironment in the vagina, conducive to maintaining the integrity of cervical epithelial and mucus barrier, protecting the host from pathogen invasions [6]. The dysbiosis of CVM, in company with changes in microbial compositions, metabolites and immune microenvironment, will damage the barrier and epithelial cells, and disrupt the immune responses against HPV infections [8], ultimately leading to the development of cervical cancer [9].

The correlation among CVM dysbiosis, cervical HPV infection and cervical cancer progression has been widely proved [10, 11], while there is still no evidence for their causal links. Furthermore, little is known about the mechanisms how CVM participates in the disease progression. Thus, this review will overview the composition of CVM, its association with HPV infection and cervical cancer development, related mechanisms, and prospects for therapeutic applications (Fig. 1).

Fig. 1
figure 1

The role of cervicovaginal microbiota (CVM) in cervical cancer (CC). The homeostasis of CVM is dominated by Lactobacillus, forming an acidic microenvironment against pathogens invasion, such as HPV infection. Affected by host, exogenous, and endogenous factors, it turns to dysbiosis, a pro-inflammatory microenvironment with anaerobes dominance. Altered microbial, metabolic, and immune signatures lead to persist HPV infection and carcinogenesis. CVM differs in HPV/CIN/CC, so specific microbial species can be used as biomarkers. Modulation of CVM can enhance therapeutic efficacy, reduce adverse reactions and improve life quality. HPV: human papilloma virus. CIN: cervical intraepithelial neoplasia

Microbiota in the female reproductive tract (FRT)

Construction and characteristics

Compared to the gastrointestinal microbiota, little is known about the role of reproductive tract microbiota in human diseases [12]. In contrast with other body sites, the reproductive tract harbors a microbiota with lower diversity, mainly dominated by Lactobacillus species [13]. Lactobacilli is vital for female reproductive health due to its probiotic activity in the microbiota [14]. Because of cervical mucus plugs, the female reproductive tract (FRT) can be divided into the lower FRT (vagina and cervix, with more microorganisms), and the upper FRT (uterus and oviduct, relatively sterile) [15]. Microbiota in lower FRT and upper FRT of the same female shows a continuity. With the rise in position, the overall biomass turns to decrease while microbial diversity gradually increases [16]. The cervicovaginal microbiota (CVM) is mainly dominated by Lactobacillus spp., while the abundance of Lactobacillus spp. in uterine microbiota (UM) is relatively lower [17].

Composition of cervicovaginal microbiota (CVM)

Generally, there is no significant difference between cervical microbiota and vaginal microbiota, jointly referred to as CVM [18]. Ravel et al. were the first to classify the CVM by microbial community structure [19] and had defined 5 community state types (CSTs). CST I, II, III, and V are dominated by a particular Lactobacillus species, respectively L. crispatus, L. gasseri, L. iners and L. jensenii. CST IV is a heterogeneous group typified by a combination of diverse facultative or strictly anaerobic bacteria and the depletion of Lactobacillus. CST IV has been subdivided into CST IV-A, IV-B, and IV-C [20]. CST IV-A has a high relative abundance of Candidatus Lachnocurva vaginae (formerly known as bacterial vaginosis-associated bacteria 1, BVAB1), while CST IV-B has a high relative abundance of Gardnerella vaginalis. Both IV-A and IV-B have moderate relative abundances of Atopobium vaginae (now reclassified as Fannyhessea vaginae [21]). CST IV-C has been divided into five sub-CSTs: CST IV-C0 is an even community with a moderate amount of Prevotella, CST IV-C1 is dominated by Streptococcus, CST IV-C2 is dominated by Enterococcus, CST IV-C3 is dominated by Bifidobacterium and CST IV-C4 is dominated by Staphylococcus (Fig. 2). Analysis of the relative abundance of bacteria revealed that the vagina and cervix showed high similarity in the microbial composition, suggesting ascending bacterial colonization from the vagina to the cervix, despite the cervical microbiota with a lower abundance of Lactobacillus and a higher abundance of Prevotella [22].

Fig. 2
figure 2

Representation of cervicovaginal bacterial community groups. CST: community state type

Sarah Lebeer et al. divided the CVM into four main modules of co-abundant cervicovaginal taxa based on compositional correlation network analyses, including L. crispatus module, Gardnerella module, Prevotella module, and Bacteroides module [23]. Some positive or negative correlations between the constituent taxa of these modules were observed, suggesting hidden interactions among the CVM components, though needed to be further experimentally validated.

Uterine microbiota (UM)

Although the upper FRT used to be considered sterile, with the application of next-generation sequencing (NGS) technologies, recent studies have recognized the presence of UM and found a possible association between UM and female reproductive health [22]. Due to the low biomass and sample contamination during collection and processing, the identification of UM compositions varies significantly across studies. However, the lower relative abundance of Lactobacillus and higher microbial diversity of UM are observed in common [22, 24, 25], significantly different from the lower FRT microbiota. Generally, the community state of UM affects fertilization and pregnancy outcomes [24, 26]. However, most samples of UM are not from healthy individuals, so the exact composition of UM and its impact on the health of women and infants remains inconclusive [25].

CVM Homeostasis and FRT health

CVM is a dynamic ecosystem in that its composition and relative abundance of bacterial species can fluctuate over short periods [27, 28]. Generally, a low microbial diversity with the dominance of Lactobacillus in CVM is considered as the homeostasis of CVM, beneficial to female reproductive health [29].

What regulates the CVM

The composition of CVM varies in different women, due to some host-related factors, such as age, menstrual cycle, pregnancy status, genetic heterogeneity, racial and ethnic differences [19, 30,31,32]. Moreover, the homeostasis of CVM can be disrupted by some exogenous factors [23, 30, 33], such as using hormonal contraceptives and smoking, both related to cervical cancer development [34]. The microorganisms in CVM may interact with each other, regulate the CVM homeostasis by themselves [23].

The state of CVM is closely associated with the level of estrogen, as estrogen promotes glycogen accumulation in vaginal epithelial cells, providing the substrate for Lactobacillus to produce lactic acids [35]. The abundance of Lactobacillus, changes dynamically during the menstrual cycle with the fluctuation of estrogen levels [30, 36,37,38]. In CVM of adolescent or postmenopausal women, relatively lack of estrogen, the microbial species richness is decreased, but species diversity is increased significantly [39, 40]. The abundance of Lactobacillus spp. decreases, while the proportion of anaerobic bacteria increases [41]. In contrary, the CVMs in pregnant women are more stable than in non-pregnant women [26, 42], with lower microbial richness and diversity due to elevated levels of estrogen and progesterone [43, 44]. However, pregnant women are still more vulnerable because the increase of other bacteria being besides higher lactobacilli loads during pregnancy [14, 45].

However, the effect of hormonal contraceptives on CVM is debated. Some studies have suggested that synthetic estrogen in the compound oral contraceptives (COCs) is beneficial for Lactobacillus dominance [46,47,48], while others found no impact on CVM [49, 50]. The CVM become more diverse in the use of only progesterone (levonorgestrel intrauterine system, LNG-IUS) [51]. Moreover, COC use is associated with increased levels of inflammatory cytokines in the cervix [39, 52]. Such an inflammatory environment may relate to carcinogenesis [53, 54].

Behavioral factors can disrupt the CVM homeostasis. Smoking can induce a higher CVM diversity and the production of biogenic amines (BAs) in vagina [55], associated with pathogen invasions and vaginal malodor [56]. Menstrual hygiene, sexual behaviors, and childbirth are correlated with the presence of bacterial vaginosis-associated bacteria (BVABs) [23].

Contribution of CVM to female reproductive health

Lactobacillus dominance

Lactobacillus is the most important component of CVM, playing an essential role in maintaining female reproductive health. Lactobacilli contribute to the reinforcement of the host immune system against several primary and opportunistic pathogens [57]. Lactobacilli produce lactic acid by fermenting glucose and maltose from vaginal epithelial cells, maintaining the vaginal pH at 3.8–4.5. Such an acidic microenvironment can inactivate pathogens and help regulate inflammatory responses [58], preventing pathogen invasions to the upper FRT [59]. Lactobacilli also secrete various metabolites that play antibacterial, antiviral, and immunomodulatory roles [60], such as bacteriocins, biosurfactants, and H2O2, inhibiting the proliferation of other microorganisms and the production of tumorigenic substances [61, 62]. The vaginal acidic environment is also beneficial to maintain the activity of bacteriocins and H2O2.

Although over 20 species of Lactobacillus have been detected in the vagina, CVM of most women is dominated by a single species of Lactobacillus, providing colonization resistance against pathogens, such as BVABs and HPVs [29]. The probiotic activity in CVM is caused not only by individual Lactobacillus species but also by its multi-microbial interaction as consortia [63]. Lactobacilli have a strong adhesion ability to the epithelial cells [64], which enables them to dominate the CVM and form a biological barrier, competing with pathogens for living space and nutrition [65]. Lactobacilli also can inhibit pathogenic adhesion and induce its displacement [66]. In addition, lactobacilli have a robust antimicrobial activity that can kill pathogens through direct contact [67].

The dominance of Lactobacillus can eliminate HPV infections, and even alleviate the progression of cervical lesions. Generally, CST I (L. crispatus dominance) and CST II (L. gasseri dominance) are the most frequent types in HPV negative women [68]. Moreover, CST II is associated with the most rapid clearance of acute HPV infection among HPV positive women [69]. Such homeostasis of CVM can be the protective factor for hrHPV clearance, with higher levels of soluble immunoglobulin A (sIgA), interleukin 2 (IL-2), and IL-1 in the cervicovaginal microenvironment [70].

Furthermore, lactobacilli and related metabolites adversely affect the growth and survival of cervical cancer cells [71]. The exopolysaccharides (EPSs), phosphorylated polysaccharides, and peptidoglycans secreted by the vaginal lactobacilli can inhibit the proliferation of cervical cancer cells and promote the process of apoptosis [72,73,74]. Studies have shown that lactobacilli and their supernatant had cytotoxic effects on cervical tumor cells but not on normal cervical epithelial cells [75], and were not affected by pH or lactic acid in the vaginal environment [76].

Bifidobacterium

Recent reports have identified the CVM dominated by Bifidobacterium in some healthy reproductive-aged women [77]. It is hypothesized that Bifidobacterium may provide a potential protective role similar to Lactobacillus in that it can also produce lactic acid and H2O2. To some extent, the proportion of Bifidobacteria and Lactobacillus may play a role in eliminating HPV infection [78]. Studies have indicated that some probiotics, such as Bifidobacterium longum, Lactobacillus johnsonii, Lactobacillus plantarum, Lactobacillus fermentum, and Lactobacillus delbrueckii, can inhibit various signaling pathways activated during HPV infection [79, 80], such as nuclear factor-κB (NF-κB) signaling pathway [81]. However, due to the scarcity of samples with Bifidobacterium dominance, more studies are needed to further clarify the clinical significance of Bifidobacterium in CVM [77].

Others

Generally, the abundances of other bacteria and fungi in the CVM, such as Atopobium and Candida albicans, are significantly lower than that of Lactobacillus, so the presence of these microorganisms usually causes no physical symptoms [82]. However, when the homeostasis of CVM is disrupted and the biological barrier formed by Lactobacillus no longer exists, these microorganisms, such as G. vaginalis and other anaerobes, will proliferate, develop biofilms, and cause recurrent female reproductive infections [83].

CVM dysbiosis and HPV infections/carcinogenesis

The different CVM composition results in different susceptibility to HPV infections. Interaction of microorganisms and metabolites with host epithelial and immune cells can alter microenvironmental signatures, ultimately affecting defense against pathogen infections and disease progression [84]. In a healthy CVM, plenty of lactobacilli can maintain the low vaginal pH and produce bacteriocin, promoting an anti-inflammatory state in the vaginal epithelium, protecting its integrity and preventing the basal cells from HPV invasion [61].

When there is dysbiosis, lactobacilli are significantly reduced or absent, replaced by specialized or facultative anaerobic bacteria [85], weakening the vaginal defense. Enzymes secreted by the dysbiotic bacterial communities, such as sialidase, can disrupt mucus barrier [86], damage cervicovaginal epithelium [87], making the basal cells vulnerable to HPV infections [4, 88]. In addition, specific toxins from the bacteria can damage host DNA, leading to the integration of viral oncogenes into host genomes [13, 89, 90].

Microbial composition and HPV persistence

The incidence and clearance rate of HPV infection varies in different CSTs [70, 91]. CST I is associated with lower HPV prevalence and higher detection rates of normal cells in cervical cytology [92]. In comparison, women with CST III and CST IV are 2–3 times more likely to be infected with hrHPV [93]. CST II (L. gasseri dominance) is associated with the fastest HPV remission rate, while CST IV-b (dominated by Fannyhessea vaginae), in contrast, shows the slowest remission rate [94]. The abundance of Lactobacillus is related to the clearance of hrHPV infection, while BVABs are linked with HPV persistence [95].

CST III and IV are believed to be less protective, associated with CVM dysbiosis, persistent HPV infection, and the development of cervical lesions [96].

CST III (L. Iners dominance)

L. iners is a transitional species that dominates the CVM after disturbance. L. iners is less able to inhibit colonization of pathogens. It can coexist with other bacteria in a wide range of pH and other metabolic stress-related situations [97, 98].

The limited protection provided by L. iners may be related to the fact that it can produce only L-lactic acid [99]. There are two isoforms of lactic acid. D-lactic acid has been reported to have a more significant inhibitory effect on exogenous bacteria than L-lactic acid [100]. L. crispatus (CST I) and L. gasseri (CST II) can produce both D- and L- lactic acid, L. jensenii (CST V) can produce only D-lactic acid, while L. iners lacks the gene that codes for D-lactate dehydrogenase (LDH) in its genome [101], resulting in a high L/D lactic acid ratio in CST III. The high L/D lactic acid ratio in vagina is related to the increase of extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-8 (MMP-8), which are known to alter the tight junctions in the endocervical epithelium, making the female genital tract susceptible to infections [102, 103].

In addition, L. iners can produce inerolysin [104], a pore-forming cholesterol-dependent cytolysin (CDC), similar to the vaginolysin produced by Gardnerella [105]. It enables L. iners to obtain nutrients from host cells by creating aqueous pores within the cell membrane [104], which may disrupt the epithelial barrier. It can be hypothesized that the dominance of L. iners offers a favorable environment for pathogens like Gardnerella to survive and destabilize the CVM [102].

In cervical cancer, tumor-resident L. iners, as an obligate L-lactate-producing lactic acid bacterium, can alter tumor metabolism and lactate signaling pathways, causing therapeutic resistance and decreased survival in patients [106]. EMMPRIN and MMP-8, increaesd by high vaginal L/D lactic acid ratio, are also involved in cancer metastasis [103].

CST IV (non-Lactobacillus dominance)

CST IV, as a combination of diverse anaerobic bacteria, is often related to CVM dysbiosis [91].

Bacterial vaginosis (BV) is the most prevalent vaginal dysbiosis, characterized by a decrease in Lactobacillus and an increase in anaerobic bacteria, such as G. vaginalis, Atopobium, and Prevotella [65], which correspond to CST IV-A, IV-B, and IV-C0. BV leads to higher vaginal pH (above 4.5), vaginal malodor and irritation, pro-inflammatory vaginal environment and increased microbial diversity in CVM [107]. The inflammation activated by BV increases the levels of some cytokines that can be related to HPV infections [108,109,110]. As a result, BV is associated with adverse reproductive health outcomes and elevated risks for STIs (especially HPVs) [111].

BVABs (such as G. vaginalis and Prevotella) may contribute to HPV infection and persistence [91, 96]. Vaginolysin secreted by G. vaginalis can cause cellular lysis and tissue breakdown [105], promoting the integration of HPV DNA into keratinocytes. Prevotella species are also reported to link with HPV infection, as they increase the microbial diversity and disturb the CVM homeostasis by providing nutrients for other BVABs [112].

Aerobic vaginitis (AV) is another vaginal dysbiosis characterized by the loss of Lactobacillus and an increase in aerobes such as Enterococcus, Escherichia coli, Staphylococcus, and Streptococcus [113, 114], which correspond to CST IV-C1, IV-C2, and IV-C4. Similarly to BV, AV can also increase the risk of HPV infections.

Altered metabolic signatures and carcinogenesis

CVM dysbiosis will alter the cervicovaginal metabolic profiles, conducive to HPV persistence [115]. A healthy metabolic microenvironment is characterized by high level of lactic acids, positively associated with the metabolism of lysolipids, phospholipids, glutathione, and glycogen, but negatively with the metabolism of biogenic amines (BAs), lysine, and histidine [116]. The Lactobacillus dominance is correlated with specific metabolites, such as anti-inflammatory nucleotides [115].

On the contrary, the metabolic microenvironment in women with BV is positively associated with BAs, lysine, and histidine metabolism, but negatively with lipid, glutathione, and glycogen metabolism. The levels of BAs (putrescine, cadaverine, and trimethylamine) and short-chain fatty acids (SCFAs) (especially acetate, butyrate, and formate) in BV are significantly high, while the levels of some amino acids (tyrosine and glutamate) in BV are relatively low [117]. In AV, the glycolytic metabolite GalNAc (N-acetylgalactosamine) and sucrose are downregulated, supporting the decrease of lactic acid [118]. The altered vaginal metabolic profiles can connect CVM dysbiosis to HPV infection and cervical carcinogenesis [115, 117].

Effects of biogenic amines (BAs)

BV is characterized by the loss of lactic acid and greater concentrations of mixed BAs (including polyamines putrescine, cadaverine, and trimethylamine) and SCFAs (including acetate, propionate, butyrate, and succinate), resulting in the higher vaginal pH and a pro-inflammatory vaginal environment [107].

Vaginal biogenic amines are the biomarker of BV, related to the vaginal malodor [119] and the reduction of Lactobacillus [120]. High production of BAs and nitrosamines leads to oxidative stress (OS) and nitrifying stress (NS). NS is associated with higher and greater pathogen resistance to the host defence systems, disturbing the immune responses [121]. OS is associated with numerous DNA lesions and protein modifications, contributing to carcinogenesis. Moreover, BAs may facilitate the formation of bacterial biofilms that entrap anaerobic bacteria, leading to their overgrowth and preventing the dominance of Lactobacillus [121].

Effects of short-chain fatty acids (SCFAs)

The effects of SCFAs are studied more in the gut than in FRT. SCFAs act as an energy source and immune modulator of the intestinal cell. Most studies show that SCFAs (especially butyrate) restore intestinal barrier function in inflammatory conditions by exhibiting anti-inflammatory effects in intestinal mucosa and inducing tight junction protein expression [122]. However, SCFAs appear to be pro-inflammatory in the FRT. BV organic acids (especially acetic and butyric acids) enhance the secretion of tumor necrosis factor α (TNF-α) after Toll-like receptor (TLR) 1/2/3 stimulation of cervicovaginal epithelial cells but inhibit the production of IL-6, RANTES (Regulated on Activation, Normal T cells Expressed and Secreted), and interferon-γ-induced protein 10 (IP-10) [123]. The difference may depend on the type and concentration of SCFAs, local pH and cell type, so the exact role of SCFAs in the FRT remains to be elucidated [124].

Altered immune signatures and carcinogenesis

CVM dysbiosis can disturb the responses of host immune system by triggering inflammations, conducive to hrHPV infections [125]. The increased diversity of CVM leads to more production of cytokines and chemokines, amplifying the inflammation, and causing cell damage [89, 92,93,94]. The dysregulated immune response can create appropriate microenvironment for persistent HPV infection [60] and tumor development [89, 126].

Dysbiotic bacterial communities and their metabolites can stimulate local immune cells, leading to production of various inflammatory cytokines and reactive oxygen species (ROS) [127]. Acute inflammation may be protective for HPV clearance [8]. However, chronic inflammation and oxidative damage by ROS can exhibit genotoxic effects on epithelial cells [127, 128], consequently leading to cell apoptosis and tumourigenesis [89]. The dysbiotic microenvironment also contributes to cell proliferation, survival and migration, and angiogenesis, all of which are hallmarks of cancer [6, 89, 129]. However, it is still unknown whether CVM dysbiosis is involved in the immune escape of HPV [128]. Moreover, the long-term effects of CVM on host immune responses against cancer cells are barely studied.

Innate immune response

The CVM plays a significant role in shaping the immune response responsible for HPV clearance [8]. The bacterial or viral components are recognized by epithelial cells through TLRs, activating the innate immune response by releasing various pro-inflammatory cytokines [87, 128]. Macrophages and dendritic cells (DCs), as antigen presenting cells (APCs), are then activated and recruit immune effector cells, such as Natural Killer (NK) cells. APCs also stimulate antigen-specific T cells and B cells to activate the adaptive immune response.

CVM dysbiosis is associated with increased pro-inflammatory cytokines that can stimulate cell proliferation and promote the development of cervical cancer [108, 109]. Studies have shown that CST IV is related to the increase of IL-1α, IL-1β, granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-10 in cervix and vagina [130]. CST III is associated with increased IP-10 and monokine induced by interferon-γ (MIG) compared with CST I/II [130].

When it comes to specific cervicovaginal microbial species, Lactobacillus usually plays a protective role in vaginal immunomodulation. Lactobacillus has the capacity to improve antiviral defenses and modulate inflammation-mediated damage [108]. Lacobacilli can promote the epithelial cells to release surfactant proteins [57]. For example, L. gasseri LGV03 can significantly increase the production of interferon α (IFN-α) and IFN-β in HPV-positive cervical epithelial cells and reduce the expression of the pro-inflammatory cytokines like IL-6, IL-8, and IL-1β [71]. Lactic acid produced by Lactobacillus can act directly on the cervicovaginal epithelium, inducing the production of the anti-inflammatory cytokine, such as IL-1Ra, and reducing pro-inflammatory cytokine production [131]. In comparison, G. vaginalis or Prevotella bivia usually induce the increase of pro-inflammatory cytokines, like IL-6, TNF-α, IL-1α, and MMP-9 [108]. Fannyhessea vaginae and Sneathia amnii elicit more robust cytokine responses, including IL-6, IL-8, IP-10, monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein 3α (MIP-3α), RANTES, MMP-10, and MMP-1 [108]. Other BVABs, such as Eggerthella, only causes an increase in IL-1α; Mobiluncus mulieris increases IL-1α, IL-6, IL-8, MCP-1, and TNF-α; while Megasphaera micronuciformis increases IL-1α, IL-1β, IL-1RA, TNF-α, and IL-6 [132].

These altered cytokines can serve as immune markers to predict BV status, HPV clearance, and CIN progression. It’s reported that high IL-1β/IP-10 ratio in BV is associated with lower rate of hrHPV clearance [110]. Elevated TNF-α/MIP-1β ratio in BV is prospectively associated with progression of persist HPV infections to CIN [110].

BVABs also stimulate the maturation and differentiation of APCs. Megasphaera elsdenii and Prevotella timonensis significantly promote the maturation of DCs, while the effects of G. vaginalis and Lactobacillus are not obvious [133]. G. vaginalis and its supernatants can induce THP-1 macrophages to differentiate into the M1 phenotype, which is involved in defence against bacterial infections, elevated ROS levels, and stimulation of the NF-κB/STAT1 (Signal Transducer and Activator of Transcription 1) pathway [134]. In contrast, vaginal Lactobacillus promotes M2 macrophages polarisation, which is involved in tissue repair and wound healing, helpful to restore the integrity of epithelial barrier [135].

The effects of bacteria on NK cells are rarely understood. NK cells are a critical component of the innate immune system, providing protection against a broad variety of viruses. Further studies are needed because the importance of NK cells in clearance of HPV-infected cells [136].

Adaptive immune response

The adaptive immune responses against HPV infection are most mediated by T cells. APCs activated by viral antigens can induce the effector CD8 + T cells targeting HPV-infected and neoplastic cells. CVM dysbiosis may induce a shift from anti-viral to anti-microbial immune response, resulting in HPV persistence [8, 137].

BVABs can act as pro-inflammatory factors, promoting the recruitment and differentiation of T cells [133]. M. elsdenii and P. timonensis significantly promote the differentiation of T cells into the pro-inflammatory Th1 type, with the increase release of IL-1β, IL-6, IL-8, IL-12p40, and TNF-α [133]. These cytokines can recruit Th1 and Th17 pro-inflammatory CD4 + T cells, effector memory CD8 + T cells, and leucocytes [138]. In contrast, Lactobacillus plays an anti-inflammatory role, promoting the differentiation of CD4 + T cells toward immunosuppressed Treg cells [139].

Reduction of lactobacilli and the less acidic environment may act as a pro-cancer factor, activating pathways related to cell proliferation and angiogenesis in the cervicovaginal epithelium [140]. In examination of immune mediators in local cervicovaginal microenvironment from women with or without cervical lesions, non-Lactobacillus dominance was associated with several pro-inflammatory (IL-36γ), chemotactic (IP10, MIP-1β and RANTES), haematopoietic (FLT 3 ligand) and adaptive immune response cytokines (IL-2, IL-4 and soluble CD40 ligand) [31]. In the cervicovaginal microenvironment of patients with cervical cancer, pro-inflammatory cytokines (IL-6, TNF-α), apoptosis-related proteins [soluble Fas receptor (sFas), sFas ligand, TRAIL (TNF-Related Apoptosis-Inducing Ligand)], growth and angiogenesis factors [hepatocyte growth factor (HGF), stem cell factor (SCF), vascular endothelial growth factor (VEGF)] and others [α-fetoprotein (AFP), osteopontin (OPN)] were elevated, positively correlated with vaginal pH, and negatively with the abundance of Lactobacillus [141].

CVM and Cervical Cancer Development

CVM and Oncogene expression

In cervical cancer, high-risk HPVs are essential for carcinogenesis and the maintenance of cancerous behavior. E6 and E7 are the main oncogenic protein of HPV, with the ability to bind and degrade tumor suppressor gene p53 and retinoblastoma protein (pRb) in infected host cells, causing cell proliferation out of control [85, 88].

The homeostasis of CVM can be disrupted by the expression of HPV oncogenes [142]. It has reported a two-way relationship between HPV infection and BV [143]. BV is a risk factor for HPV infection, and HPV infections are considered a cause for increased diversity, altered composition, and disordered function of CVM in turn [144]. Compared with HPV negative individuals, it is more likely to detect Pseudomonas [16], Atopobium [94, 96], Fusobacterium [92], and Sneathia [112] in CVM of HPV positive patients. Products of HPV E7 oncogene can significantly inhibit the expression of host defense peptides in the vagina (including HβD1, 2, 4, HD-5/6, SLPI, S100A7, and elafin) [143]. These peptides have antimicrobial activity against BVABs like G. vaginalis. Meanwhile, S100A7 and elafin expressed by the cervicovaginal squamous epithelial cells can be used as amino acid sources by lactobacilli for survival. Therefore, the survival of Lactobacillus species are considerably inhibited by HPVs, resulting in the CVM dysbiosis [143].

In turn, the expression of HPV oncogenes can also be regulated by the CVM in different stages of cervical lesions [85, 145]. The production of HPV oncoproteins is significantly upregulated during the progression of CIN. Along with the precancerous lesion development, the severity of CVM dysbiosis increased, including the increase of CVM diversity and richness, and the decrease of Lactobacillus [91, 146]. Various aerobic and anaerobic bacteria can be detected in the CVM with cervical lesions, such as Gardnerella vaginalis, Prevotella bivia, Sneathia sanguinegens, Megasphaera micronuciformis, and Peptostreptococcus anaerobes [147]. The expression of the HPV oncogenes is positively correlated with the presence of these microorganisms, but negatively with the presence of Lactobacillus [145]. Lactobacilli are reported to decrease the oncogene expression in cervical cancer cells, while G. vaginalis and M. micronuciformis can induce the production of viral oncoproteins [85].

CVM as Biomarker for CIN and CC Detection

The microbial composition and abundance of specific species in CVM vary during cervical cancer development, which can be used to distinguish patients with HPV infection, CIN or CC. In a study with 5 groups [healthy, HPV positive (HPV+), low-grade SIL (LSIL), high-grade SIL (HSIL), and CC], the CC group showed the highest CVM diversity, significantly different from other groups [144]. The increase of the proportion of Bacillus and Anaerococcus and the decrease of the abundance of G. vaginalis may be related to the progression of CIN [144]. Another study showed that the abundance of Gardnerella was positively correlated with the CIN progression by inducing an increased CVM diversity over time, not directly causing HSIL [95]. An increased abundance of Gardnerella is not indicative of being pathogenic, but rather reflective of different bacterial relationships and host states [23]. Higher levels of Sneathia sanguinegens, Anaerococcus tetradius, and Peptostreptococcus anaerobes and lower levels of L. jensenii can be detected in CVM of women with HSIL than that with LSIL [147]. Comparing the CIN and CC groups, the presence of Gardnerella and Streptococcus differed significantly, with the former dominant in the CIN group and the latter dominant in the CC group [148]. The abundance of Fusobacterium and Sneathia is significantly higher in advanced CC than in the early stages, and Fusobacterium necrophorum is observed only in CC [92, 112]. The presence of Fusobacterium in CVM may lead to elevated cervical expression of anti-inflammatory IL-4 and transforming growth factor β1(TGFβ1), contributing to carcinogenesis [92].

Microbial species mentioned above can be potential biomarkers for HPV infection, CIN, and CC. However, the clinical significance of these biomarkers is limited because of the small sample sizes and no assessment of environmental factors, needed to be verified in further studies [70].

CVM modulation and cancer treatment

The CVM homeostasis, with Lactobacillus dominance, is essential for female reproductive health. Besides its protective role in cervical cancer prevention, CVM can affect host responses to cancer treatment.

Treatment for cervical cancer, including surgery, radiotherapy, chemotherapy or a combination of chemoradiation, depends on the stage. Immunotherapy is a new option approved by the US Food and Drug Administration (FDA) [5]. For instance, the immune checkpoint inhibitor targeting programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1) are used in advanced cervical cancer with progression during or after chemotherapy [5].

Therapies against cancer can disturb the state of CVM [149], inducing treatment-resistance [106]. Thus, the modulation of CVM, turning the dysbiosis back to homeostasis, will improve the therapeutic efficacy, providing a potential direction for cervical cancer treatment.

Surgery

The state of CVM may be related to the recurrence of CIN or HPV infection after surgery. Effect of CIN excision on CVM is controversial. Some studies have observed a decreased CVM diversity and an increase in Lactobacillus after excision [150, 151]. However, another study reported no significant difference in CVM composition after treatment [152]. Further studies are needed to verify the association among the CVM state, CIN resection, and recurrence rates after treatment. The alternation of CVM before and after surgery in patients with cervical cancer, especially with or without the recovery of CVM homeostasis, can be essential to distinguish patients with high risk of recurrence.

Radiotherapy

Radiation can disrupt the CVM communities [153, 154]. Women with gynecological cancer before and after radiotherapy showed an increase in Mobiluncus, Atopobium, and Prevotella and a decrease in Gardnerella and Peptostreptococcus, as well as Lactobacillus following treatment [153]. The microbial diversity of CVM in cancer patients also increased after radiotherapy [154]. Radiotherapy also causes epithelium damage, resulting in various adverse effects, such as vulvovaginal atrophy (VVA), vaginal stenosis, and pelvic pain [155]. A reduced amount of Lactobacillus is observed in patients with radiation-induced VVA [156]. In addition, the presence of certain microbial species, like L. iners, was reported to induce chemoradiation resistance, while L. crispatus did not [106]. Thus, modulation of CVM may relieve related adverse reactions and reduce radiation-resistance.

Chemotherapy

Regarding chemotherapy, a study has found lower CVM diversity in CC patients can be associated with more significant responses to platinum drugs than non-responders [149], suggesting that modulation of CVM may enhance the chemotherapeutic efficacy, despite lack of exploration for mechanisms. In addition, studies on the gut microbiome reveal that microbiota can reduce the toxicity of chemotherapeutic agents and improve the efficacy of chemotherapy and immunotherapy [157].

Immunotherapy

The role of CVM in immunotherapies is still unclear. However, studies show that the diversity and composition of the gut microbiome can affect anti-tumour immunity and the efficacy of PD-1 immunotherapy in various tumor types [158,159,160]. The mechanisms include translocation, immunomodulation, drug metabolism, and enzymatic degradation [157, 161]. Moreover, studies on gut microbiomes suggest that the interactions between bacteria and the host immune system may also play roles in patient responsiveness to immunotherapeutic agents [157,158,159,160, 162,163,164]. Structural and metabolic features of microbiota can regulate the immune response to cancer cells [165,166,167].

As HPVs are essential for carcinogenesis in cervical cancer, cancer vaccine targeting HPV is a kind of immunotherapy specific to cervical cancer. Lactobacillus can not only convey antigens as a vaccine carrier, but also enhance the immune response as a vaccine adjuvant. A Lactobacillus-based oral vaccine, expressing HPV E7 protein on the surface of Lactobacillus casei strain, is undergoing clinical trials, and has induced the regression of CIN [168].

CVM Modulation

Microbiome-modulation can reduce the therapeutic toxicities, enhance the therapeutic efficacy and improve life quality for patients [157]. It also has essential health benefits in preventing the genital inflammations, STIs, and cancers.

Exogenous lactobacilli supplementation can somewhat reverse the dysregulation of CVM [169, 170]. Oral administration of a pertinent lactobacilli strain mixture can improve vaginal health in asymptomatic women with vaginal dysbiosis [171]. Furthermore, probiotics consisting of Lactobacillus spp. may increase the clearance of HPV and delay the progression of cervical cancer [172]. Long-term (6 months) use of Lactobacillus rhamnosus BMX 54 is twice as likely to resolve HPV-associated cytological abnormalities than short-term use (3 months) [173].

Moreover, the feasibility of vaginal microbiome transplantation (VMT) as the treatment for women with vaginal disorders has been proven [174]. However, the long-term effects remain unknown, and the potential risks remain elusive.

Discussion

With the advance of sequencing technology, the composition of FRT microbial communities, specific bacterial species, and their contributions to health and disease have been preliminarily identified. However, the 16S rRNA sequencing technique still has some limitations, especially a lack of absolute bacterial quantification. Other molecular techniques for bacterial quantification, for example, quantitative real-time PCR or flow cytometry, could be used to determine absolute loads of specific bacteria associated with particular conditions [13, 110].

CVM is dynamic, and various factors can regulate its constructure and composition [30]. Some women with CVM abnormality complain no apparent symptoms. It is difficult to distinguish whether such CVM alteration is a normal fluctuation or an indication of female reproductive diseases. Longitudinal studies are required to evaluate the necessity of intervention.

Besides Lactobacillus, some studies have reported the dominance of other lactic acid bacteria in the CVM [77, 175]. However, whether these bacteria can provide the similar protective effects to Lactobacillus, such as the colonization resistance against pathogens, still remains to be explored.

The impacts of exogenous factors related to cervical cancer on CVM, like hormonal contraceptives and smoking, are controversial. It is hard to tell whether they increase the risk of CC by affecting CVM or promote the genesis of CC resulting in changes in CVM [56].

The development of cervical cancer is closely related to HPV infection. Although HPV infections, in general, are non-inflammatory, inflammatory reactions induced by CVM dysbiosis have been proven contributing to the development of CIN. Moreover, the long-term persistence of HPV is thought to result in generalized immunosuppression [121]. However, studies have reported that some BVABs, such as G. vaginalis, are involved in a shift from antimicrobial to antiviral responses, related to HPV clearance [8, 87]. In contrast, Lactobacillus plays an immunosuppressive role [139]. It is still unknown how CVM dysbiosis involves in the immune escape of HPV [128], and long-term effects of CVM on host immune responses against cancer cells are barely studied [121]. Notably, Gardnerella species are now divided into Gardnerella vaginalis, Gardnerella leopoldii, Gardnerella piotii, and Gardnerella swidsinskii, as these different species show distinct ecological or pathological properties [176]. The disputed pathogenic role of G. vaginalis may be the result of formerly considering different species as a single species.

Studies about mechanisms of CVM in cervical carcinogenesis are relatively rare. Integrated multi-omics approaches have been used to identify microbial and host signatures (bacterial communities and species, immune mediators and other proteins, and metabolites) in the cervicovaginal microenvironment. 3D cell cultures and mouse models are required in further research to determine the mechanism for the role of microbial communities or single specific microbial species with host-microbiota interactions in cervical carcinogenesis [108, 177].

Most studies about microbiota and carcinogenesis are focused on the gut microbiota. However, there are significant differences between the two body sites. There is an acidic environment in the vagina (pH < 4.5), while the pH of the intestinal environment is over 7.0. Besides, a healthy CVM is associated with low diversity, while in the intestine, high microbial diversity is considered a sign of health [178]. What’s more, the concentration and composition of microbial metabolites are different, leading to different effects on cells, which also differ depending on pH and cell types. In addition, considering that the CVM can be regulated by estrogen levels, and that circulating estrogen levels in the body can be influenced by the gut microbiota, there may be a connection between the gut and cervicovaginal microbiomes [156, 179]. The modulation of gut microbiome may contribute to the restoration of CVM homeostasis.

The interactions between CVM and cancer treatment have not been thoroughly studied. More studies are needed to verify the therapeutic efficacy affected by CVM and the recurrence rate of gynecological diseases after modulation of CVM.

Conclusion

CVM is different from microbial communities in other body sites, with a low diversity and mainly dominated by Lactobacillus. However, its role in cervical cancer development and related mechanisms remains unclear. Homeostasis of CVM is crucial for maintaining female reproductive health, and such a dynamic ecosystem can be affected by host, exogenous, and endogenous factors. Dysbiosis of CVM, including changes in microbial, metabolic, and immune signatures, can form a pro-inflammatory microenvironment, weaken the resistance to pathogens (including HPVs), and contribute to carcinogenesis. Numerous studies have demonstrated the association of cervicovaginal dysbiosis with HPV infection, CIN, and CC. Besides, specific microbial species have been identified as biomarkers for HPV/CIN/CC. Several studies have explored the mechanisms of microbial interactions with HPV and cancer cells, but further research is still needed to confirm the influence of microbiota on disease in 3D cell cultures and mouse models. Cancer treatment can also affect CVM, and modulation of CVM can help enhance therapeutic efficacy, alleviate adverse reactions, and improve the life quality of patients. Understanding the role of CVM in cervical cancer development may provide new opportunities for cancer prevention, treatment, and improvement of female life quality and overall health.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. https://doi.org/10.3322/caac.21660.

    Article  CAS  PubMed  Google Scholar 

  2. Singh D, Vignat J, Lorenzoni V, et al. Global estimates of incidence and mortality of cervical cancer in 2020: a baseline analysis of the WHO Global Cervical Cancer Elimination Initiative. Lancet Glob Health. 2023;11(2):e197–206. https://doi.org/10.1016/S2214-109X(22)00501-0.

    Article  CAS  PubMed  Google Scholar 

  3. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12–49. https://doi.org/10.3322/caac.21820.

    Article  PubMed  Google Scholar 

  4. Cascardi E, Cazzato G, Daniele A, et al. Association between Cervical Microbiota and HPV: could this be the Key to complete. Cerv Cancer Eradication? Biology. 2022;11(8):1114. https://doi.org/10.3390/biology11081114.

    Article  CAS  Google Scholar 

  5. Cohen PA, Jhingran A, Oaknin A, et al. Cervical cancer. Lancet. 2019;393(10167):169–82. https://doi.org/10.1016/S0140-6736(18)32470-X.

    Article  PubMed  Google Scholar 

  6. Trifanescu OG, Trifanescu RA, Mitrica RI, et al. The Female Reproductive Tract Microbiome and Cancerogenesis: a Review story of Bacteria, hormones, and Disease. Diagnostics (Basel). 2023;13(5):877. https://doi.org/10.3390/diagnostics13050877.

    Article  CAS  PubMed  Google Scholar 

  7. Salinas AM, Osorio VG, Pacha-Herrera D, et al. Vaginal microbiota evaluation and prevalence of key pathogens in Ecuadorian women: an epidemiologic analysis. Sci Rep. 2020;10(1):18358. https://doi.org/10.1038/s41598-020-74655-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Moscicki AB, Shi B, Huang H, et al. Cervical-vaginal Microbiome and Associated Cytokine profiles in a prospective study of HPV 16 Acquisition, persistence, and Clearance. Front Cell Infect Microbiol. 2020;10:569022. https://doi.org/10.3389/fcimb.2020.569022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang Y, Wang X, Zhu M, et al. The interplay between Cervicovaginal Microbial Dysbiosis and Cervicovaginal Immunity. Front Immunol. 2022;13:857299. https://doi.org/10.3389/fimmu.2022.857299.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhai Q, Zhang W, Zhang Z, et al. Characteristics of the Cervicovaginal Microenvironment in Childbearing-Age women with different degrees of cervical lesions and HR-HPV positivity. Pol J Microbiol. 2021;70(4):489–500. https://doi.org/10.33073/pjm-2021-046.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Wei B, Chen Y, Lu T, et al. Correlation between vaginal microbiota and different progression stages of cervical cancer. Genet Mol Biol. 2022;45(2):e20200450. https://doi.org/10.1590/1678-4685-gmb-2020-0450.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Xia C, Su J, Liu C et al. Human microbiomes in cancer development and therapy. MedComm (2020). 2023;4(2):e221. https://doi.org/10.1002/mco2.221.

  13. Laniewski P, Ilhan ZE, Herbst-Kralovetz MM. The microbiome and gynaecological cancer development, prevention and therapy. Nat Rev Urol. 2020;17(4):232–50. https://doi.org/10.1038/s41585-020-0286-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Pacha-Herrera D, Vasco G, Cruz-Betancourt C, et al. Vaginal microbiota evaluation and Lactobacilli quantification by qPCR in pregnant and non-pregnant women: a pilot study. Front Cell Infect Microbiol. 2020;10:303. https://doi.org/10.3389/fcimb.2020.00303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dong M, Dong Y, Bai J, et al. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol. 2023;13:1124591. https://doi.org/10.3389/fcimb.2023.1124591.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhang Z, Li T, Zhang D, et al. Distinction between vaginal and cervical microbiota in high-risk human papilloma virus-infected women in China. BMC Microbiol. 2021;21(1):90. https://doi.org/10.1186/s12866-021-02152-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang R, Zhou G, Wu L, et al. The Microbial composition of Lower Genital Tract May affect the outcome of in vitro fertilization-embryo transfer. Front Microbiol. 2021;12:729744. https://doi.org/10.3389/fmicb.2021.729744.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Anahtar MN, Gootenberg DB, Mitchell CM, et al. Cervicovaginal Microbiota and Reproductive Health: the Virtue of simplicity. Cell Host Microbe. 2018;23(2):159–68. https://doi.org/10.1016/j.chom.2018.01.013.

    Article  CAS  PubMed  Google Scholar 

  19. Ravel J, Gajer P, Abdo Z, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci. 2011;108(supplement1):4680–7. https://doi.org/10.1073/pnas.1002611107.

    Article  PubMed  Google Scholar 

  20. France MT, Ma B, Gajer P, et al. VALENCIA: a nearest centroid classification method for vaginal microbial communities based on composition. Microbiome. 2020;8(1):166. https://doi.org/10.1186/s40168-020-00934-6.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Nouioui I, Carro L, García-López M, et al. Genome-based taxonomic classification of the Phylum Actinobacteria. Front Microbiol. 2018;9:2007. https://doi.org/10.3389/fmicb.2018.02007.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Sun N, Ding H, Yu H, et al. Comprehensive characterization of Microbial Community in the female genital tract of Reproductive-aged women in China. Front Cell Infect Microbiol. 2021;11:649067. https://doi.org/10.3389/fcimb.2021.649067.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lebeer S, Ahannach S, Gehrmann T, et al. A citizen-science-enabled catalogue of the vaginal microbiome and associated factors. Nat Microbiol. 2023;8(11):2183–95. https://doi.org/10.1038/s41564-023-01500-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Baker JM, Chase DM, Herbst-Kralovetz MM. Uterine microbiota: residents, tourists, or invaders? Front Immunol. 2018;9:208. https://doi.org/10.3389/fimmu.2018.00208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Winters AD, Romero R, Gervasi MT, et al. Does the endometrial cavity have a molecular microbial signature? Sci Rep. 2019;9(1):9905. https://doi.org/10.1038/s41598-019-46173-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shen L, Wang W, Hou W, et al. The function and mechanism of action of uterine microecology in pregnancy immunity and its complications. Front Cell Infect Microbiol. 2023;12:1025714. https://doi.org/10.3389/fcimb.2022.1025714.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kaur H, Merchant M, Haque MM, et al. Crosstalk between Female Gonadal Hormones and vaginal microbiota across various phases of women’s gynecological lifecycle. Front Microbiol. 2020;11:551. https://doi.org/10.3389/fmicb.2020.00551.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Gajer P, Brotman RM, Bai G, et al. Temporal dynamics of the human vaginal microbiota. Sci Transl Med. 2012;4(132):132ra52. https://doi.org/10.1126/scitranslmed.3003605.

    Article  PubMed  PubMed Central  Google Scholar 

  29. McKinnon LR, Achilles SL, Bradshaw CS, et al. The evolving facets of bacterial vaginosis: implications for HIV Transmission. AIDS Res Hum Retroviruses. 2019;35(3):219–28. https://doi.org/10.1089/aid.2018.0304.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Song SD, Acharya KD, Zhu JE, et al. Daily Vaginal Microbiota fluctuations Associated with Natural Hormonal cycle, contraceptives, Diet, and Exercise. mSphere. 2020;5(4):e520–93. https://doi.org/10.1128/mSphere.00593-20.

    Article  Google Scholar 

  31. Łaniewski P, Barnes D, Goulder A, et al. Linking cervicovaginal immune signatures, HPV and microbiota composition in cervical carcinogenesis in non-hispanic and hispanic women. Sci Rep. 2018;8(1):7593. https://doi.org/10.1038/s41598-018-25879-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Onywera H, Williamson A, Mbulawa ZZA, et al. Factors associated with the composition and diversity of the cervical microbiota of reproductive-age black South African women: a retrospective cross-sectional study. PeerJ. 2019;7:e7488. https://doi.org/10.7717/peerj.7488.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Moosa Y, Kwon D, de Oliveira T, et al. Determinants of vaginal microbiota composition. Front Cell Infect Microbiol. 2020;10:467. https://doi.org/10.3389/fcimb.2020.00467.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Kyrgiou M, Mitra A, Moscicki A. Does the vaginal microbiota play a role in the development of cervical cancer? Transl Res. 2017;179:168–82. https://doi.org/10.1016/j.trsl.2016.07.004.

    Article  PubMed  Google Scholar 

  35. Amabebe E, Anumba DOC. The Vaginal Microenvironment: the physiologic role of Lactobacilli. Front Med (Lausanne). 2018;5:181. https://doi.org/10.3389/fmed.2018.00181.

    Article  PubMed  Google Scholar 

  36. Krog MC, Hugerth LW, Fransson E, et al. The healthy female microbiome across body sites: effect of hormonal contraceptives and the menstrual cycle. Hum Reprod. 2022;37(7):1525–43. https://doi.org/10.1093/humrep/deac094.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Oerlemans E, Ahannach S, Wittouck S, et al. Impacts of Menstruation, Community Type, and an oral yeast Probiotic on the vaginal microbiome. mSphere. 2022;7(5):e222–39. https://doi.org/10.1128/msphere.00239-22.

    Article  CAS  Google Scholar 

  38. Jie Z, Chen C, Hao L, et al. Life history recorded in the Vagino-cervical Microbiome along with multi-omes. Genomics Proteom Bioinf. 2022;20(2):304–21. https://doi.org/10.1016/j.gpb.2021.01.005.

    Article  CAS  Google Scholar 

  39. Balle C, Konstantinus IN, Jaumdally SZ, et al. Hormonal contraception alters vaginal microbiota and cytokines in South African adolescents in a randomized trial. Nat Commun. 2020;11(1):5578. https://doi.org/10.1038/s41467-020-19382-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim S, Seo H, Rahim MA et al. Changes in the Microbiome of Vaginal Fluid after Menopause in Korean Women. J Microbiol Biotechnol. 20218;31(11):1490–500. https://doi.org/10.4014/jmb.2106.06022.

  41. Gliniewicz K, Schneider GM, Ridenhour BJ, et al. Comparison of the Vaginal microbiomes of Premenopausal and Postmenopausal women. Front Microbiol. 2019;10:193. https://doi.org/10.3389/fmicb.2019.00193.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Romero R, Hassan SS, Gajer P, 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. https://doi.org/10.1186/2049-2618-2-4.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Freitas AC, Chaban B, Bocking A, et al. The vaginal microbiome of pregnant women is less rich and diverse, with lower prevalence of Mollicutes, compared to non-pregnant women. Sci Rep. 2017;7(1):9212. https://doi.org/10.1038/s41598-017-07790-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gupta P, Singh MP, Goyal K. Diversity of vaginal microbiome in pregnancy: deciphering the Obscurity. Front Public Health. 2020;8:326. https://doi.org/10.3389/fpubh.2020.00326.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Salinas AM, Osorio VG, Endara PF, et al. Bacterial identification of the vaginal microbiota in Ecuadorian pregnant teenagers: an exploratory analysis. PeerJ. 2018;6:e4317. https://doi.org/10.7717/peerj.4317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Brooks JP, Edwards DJ, Blithe DL, 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. https://doi.org/10.1016/j.contraception.2016.11.006.

    Article  CAS  PubMed  Google Scholar 

  47. Vodstrcil LA, Plummer ME, Fairley CK, et al. Combined oral contraceptive pill-exposure alone does not reduce the risk of bacterial vaginosis recurrence in a pilot randomised controlled trial. Sci Rep. 2019;9(1):3555. https://doi.org/10.1038/s41598-019-39879-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tuddenham S, Gajer P, Burke AE, et al. Lactobacillus-dominance and rapid stabilization of vaginal microbiota in combined oral contraceptive pill users examined through a longitudinal cohort study with frequent vaginal sampling over two years. EBioMedicine. 2023;87:104407. https://doi.org/10.1016/j.ebiom.2022.104407.

    Article  CAS  PubMed  Google Scholar 

  49. Donders G, Bellen G, Janssens D, et al. Influence of contraceptive choice on vaginal bacterial and fungal microflora. Eur J Clin Microbiol Infect Dis. 2017;36(1):43–8. https://doi.org/10.1007/s10096-016-2768-8.

    Article  CAS  PubMed  Google Scholar 

  50. Achilles SL, Hillier SL. The complexity of contraceptives. AIDS. 2013;27(Supplement 1):S5–15. https://doi.org/10.1097/QAD.0000000000000058.

    Article  PubMed  Google Scholar 

  51. Achilles SL, Austin MN, Meyn LA, et al. Impact of contraceptive initiation on vaginal microbiota. Am J Obstet Gynecol. 2018;218(6):621–2. https://doi.org/10.1016/j.ajog.2018.02.017.

    Article  CAS  Google Scholar 

  52. Fichorova RN, Chen P, Morrison CS, et al. The contribution of Cervicovaginal Infections to the Immunomodulatory effects of Hormonal Contraception. mBio. 2015;6(5):e215–21. https://doi.org/10.1128/mBio.00221-15.

    Article  CAS  Google Scholar 

  53. Tamma R, Annese T, Ruggieri S, et al. Inflammatory cells infiltrate and angiogenesis in locally advanced and metastatic cholangiocarcinoma. Eur J Clin Invest. 2019;49(5):e13087. https://doi.org/10.1111/eci.13087.

    Article  CAS  PubMed  Google Scholar 

  54. Yang X, Guo Y, Chen C, et al. Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology. 2021;164(3):476–93. https://doi.org/10.1111/imm.13397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Brotman RM, He X, Gajer P, et al. Association between cigarette smoking and the vaginal microbiota: a pilot study. BMC Infect Dis. 2014;14:471. https://doi.org/10.1186/1471-2334-14-471.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Nelson TM, Borgogna JC, Michalek RD, et al. Cigarette smoking is associated with an altered vaginal tract metabolomic profile. Sci Rep. 2018;8(1):852. https://doi.org/10.1038/s41598-017-14943-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cangui-Panchi SP, Ñacato-Toapanta AL, Enríquez-Martínez LJ, et al. Battle royale: Immune response on biofilms - host-pathogen interactions. Curr Res Immunol. 2023;4:100057. https://doi.org/10.1016/j.crimmu.2023.100057.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Scillato M, Spitale A, Mongelli G, et al. Antimicrobial properties of Lactobacillus cell-free supernatants against multidrug-resistant urogenital pathogens. Microbiologyopen. 2021;10(2):e1173. https://doi.org/10.1002/mbo3.1173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tamarelle J, Thiébaut ACM, de Barbeyrac B, et al. The vaginal microbiota and its association with human papillomavirus, Chlamydia trachomatis, Neisseria gonorrhoeae and Mycoplasma genitalium infections: a systematic review and meta-analysis. Clin Microbiol Infect. 2019;25(1):35–47. https://doi.org/10.1016/j.cmi.2018.04.019.

    Article  CAS  PubMed  Google Scholar 

  60. Kalia N, Singh J, Kaur M. Microbiota in vaginal health and pathogenesis of recurrent vulvovaginal infections: a critical review. Ann Clin Microbiol Antimicrob. 2020;19(1):5. https://doi.org/10.1186/s12941-020-0347-4.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Yang X, Da M, Zhang W, et al. Role of Lactobacillus in cervical cancer. Cancer Manag Res. 2018;10:1219–29. https://doi.org/10.2147/CMAR.S165228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. De Gregorio PR, Parolin C, Abruzzo A, et al. Biosurfactant from vaginal Lactobacillus crispatus BC1 as a promising agent to interfere with Candida adhesion. Microb Cell Fact. 2020;19(1):133. https://doi.org/10.1186/s12934-020-01390-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Pacha-Herrera D, Erazo-Garcia MP, Cueva DF, et al. Clustering analysis of the Multi-microbial Consortium by Lactobacillus species against vaginal dysbiosis among Ecuadorian women. Front Cell Infect Microbiol. 2022;12:863208. https://doi.org/10.3389/fcimb.2022.863208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Jung H, Ehlers MM, Peters RPH, et al. Growth forms of Gardnerella spp. and Lactobacillus spp. on vaginal cells. Front Cell Infect Microbiol. 2020;10:71. https://doi.org/10.3389/fcimb.2020.00071.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Pendharkar S, Skafte-Holm A, Simsek G, et al. Lactobacilli and their probiotic effects in the Vagina of Reproductive Age women. Microorganisms. 2023;11(3):636. https://doi.org/10.3390/microorganisms11030636.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Rodríguez-Arias RJ, Guachi-Álvarez BO, Montalvo-Vivero DE, et al. Lactobacilli displacement and Candida albicans inhibition on initial adhesion assays: a probiotic analysis. BMC Res Notes. 2022;15(1):239. https://doi.org/10.1186/s13104-022-06114-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Atassi F, Pho Viet Ahn DL, Lievin-Le Moal V. Diverse expression of Antimicrobial Activities against Bacterial Vaginosis and urinary tract infection pathogens by Cervicovaginal Microbiota strains of Lactobacillus gasseri and Lactobacillus crispatus. Front Microbiol. 2019;10:2900. https://doi.org/10.3389/fmicb.2019.02900.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Chee W, Chew SY, Than L. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb Cell Fact. 2020;19(1):203. https://doi.org/10.1186/s12934-020-01464-4.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Shannon B, Yi TJ, Perusini S, et al. Association of HPV infection and clearance with cervicovaginal immunology and the vaginal microbiota. Mucosal Immunol. 2017;10(5):1310–9. https://doi.org/10.1038/mi.2016.129.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Han M, Wang N, Han W, et al. Vaginal and tumor microbiomes in gynecological cancer (review). Oncol Lett. 2023;25(4):153. https://doi.org/10.3892/ol.2023.13739.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Gao Q, Fan T, Luo S, et al. Lactobacillus gasseri LGV03 isolated from the cervico-vagina of HPV-cleared women modulates epithelial innate immune responses and suppresses the growth of HPV-positive human cervical cancer cells. Transl Oncol. 2023;35:101714. https://doi.org/10.1016/j.tranon.2023.101714.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Sungur T, Aslim B, Karaaslan C, et al. Impact of Exopolysaccharides (EPSs) of Lactobacillus gasseri strains isolated from human vagina on cervical tumor cells (HeLa). Anaerobe. 2017;47:137–44. https://doi.org/10.1016/j.anaerobe.2017.05.013.

    Article  PubMed  Google Scholar 

  73. Zadravec P, Štrukelj B, Berlec A. Improvement of LysM-Mediated Surface Display of designed ankyrin repeat proteins (DARPins) in recombinant and nonrecombinant strains of Lactococcus lactis and Lactobacillus species. Appl Environ Microbiol. 2015;81(6):2098–106. https://doi.org/10.1128/AEM.03694-14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Homburg C, Bommer M, Wuttge S, et al. Inducer exclusion in Firmicutes: insights into the regulation of a carbohydrate ATP binding cassette transporter from Lactobacillus casei BL23 by the signal transducing protein P-Ser46-HPr. Mol Microbiol. 2017;105(1):25–45. https://doi.org/10.1111/mmi.13680.

    Article  CAS  PubMed  Google Scholar 

  75. Wang K, Xu D, Wang B, et al. Inhibitory effect of vaginal Lactobacillus supernatants on cervical Cancer cells. Probiotics Antimicrob Proteins. 2018;10(2):236–42. https://doi.org/10.1007/s12602-017-9339-x.

    Article  PubMed  Google Scholar 

  76. Motevaseli E, Shirzad M, Akrami SM, et al. Normal and tumour cervical cells respond differently to vaginal lactobacilli, independent of pH and lactate. J Med Microbiol. 2013;62(7):1065–72. https://doi.org/10.1099/jmm.0.057521-0.

    Article  PubMed  Google Scholar 

  77. Freitas AC, Hill JE. Quantification, isolation and characterization of Bifidobacterium from the vaginal microbiomes of reproductive aged women. Anaerobe. 2017;47:145–56. https://doi.org/10.1016/j.anaerobe.2017.05.012.

    Article  PubMed  Google Scholar 

  78. Mei L, Wang T, Chen Y, et al. Dysbiosis of vaginal microbiota associated with persistent high-risk human papilloma virus infection. J Transl Med. 20223;20(1):12. https://doi.org/10.1186/s12967-021-03201-w.

  79. Kim WG, Kim HI, Kwon EK, et al. Lactobacillus plantarum LC27 and Bifidobacterium longum LC67 mitigate alcoholic steatosis in mice by inhibiting LPS-mediated NF-kappaB activation through restoration of the disturbed gut microbiota. Food Funct. 2018;9(8):4255–65. https://doi.org/10.1039/c8fo00252e.

    Article  CAS  PubMed  Google Scholar 

  80. Kim DE, Kim JK, Han SK, et al. Lactobacillus plantarum NK3 and Bifidobacterium longum NK49 alleviate bacterial vaginosis and osteoporosis in mice by suppressing NF-κB-Linked TNF-α expression. J Med Food. 2019;22(10):1022–31. https://doi.org/10.1089/jmf.2019.4419.

    Article  CAS  PubMed  Google Scholar 

  81. Tilborghs S, Corthouts J, Verhoeven Y, et al. The role of Nuclear factor-kappa B signaling in human cervical cancer. Crit Rev Oncol Hematol. 2017;120:141–50. https://doi.org/10.1016/j.critrevonc.2017.11.001.

    Article  PubMed  Google Scholar 

  82. Esposito MM, Patsakos S, Borruso L. The role of the mycobiome in women’s Health. J Fungi (Basel). 2023;9(3):348. https://doi.org/10.3390/jof9030348.

    Article  PubMed  Google Scholar 

  83. Machado A, Cerca N. Influence of Biofilm formation by Gardnerella vaginalis and other Anaerobes on bacterial vaginosis. J Infect Dis. 2015;212(12):1856–61. https://doi.org/10.1093/infdis/jiv338.

    Article  CAS  PubMed  Google Scholar 

  84. Smith SB, Ravel J. The vaginal microbiota, host defence and reproductive physiology. J Physiol. 2017;595(2):451–63. https://doi.org/10.1113/JP271694.

    Article  CAS  PubMed  Google Scholar 

  85. Nicolò S, Antonelli A, Tanturli M, et al. Bacterial species from vaginal microbiota differently affect the production of the E6 and E7 oncoproteins and of p53 and p-Rb oncosuppressors in HPV16-Infected cells. Int J Mol Sci. 2023;24(8). https://doi.org/10.3390/ijms24087173.

  86. Kyrgiou M, Moscicki AB. Vaginal microbiome and cervical cancer. Semin Cancer Biol. 2022;86(Pt 3):189–98. https://doi.org/10.1016/j.semcancer.2022.03.005.

    Article  CAS  PubMed  Google Scholar 

  87. Anton L, Ferguson B, Friedman ES, et al. Gardnerella vaginalis alters cervicovaginal epithelial cell function through microbe-specific immune responses. Microbiome. 2022;10(1):119. https://doi.org/10.1186/s40168-022-01317-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Kurnia I, Rauf S, Hatta M, et al. Molecular patho-mechanisms of cervical cancer (MMP1). Ann Med Surg (Lond). 2022;77:103415. https://doi.org/10.1016/j.amsu.2022.103415.

    Article  PubMed  Google Scholar 

  89. Rajagopala SV, Vashee S, Oldfield LM, et al. The human microbiome and Cancer. Cancer Prev Res (Phila). 2017;10(4):226–34. https://doi.org/10.1158/1940-6207.CAPR-16-0249.

    Article  PubMed  Google Scholar 

  90. Piyathilake CJ, Ollberding NJ, Kumar R, et al. Cervical Microbiota Associated with higher Grade Cervical Intraepithelial Neoplasia in Women infected with high-risk human papillomaviruses. Cancer Prev Res (Phila). 2016;9(5):357–66. https://doi.org/10.1158/1940-6207.CAPR-15-0350.

    Article  CAS  PubMed  Google Scholar 

  91. Molina MA, Andralojc KM, Huynen MA, et al. In-depth insights into cervicovaginal microbial communities and hrHPV infections using high-resolution microbiome profiling. NPJ Biofilms Microbiomes. 2022;8(1):75. https://doi.org/10.1038/s41522-022-00336-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Audirac-Chalifour A, Torres-Poveda K, Bahena-Román M, et al. Cervical Microbiome and Cytokine Profile at various stages of cervical Cancer: a pilot study. PLoS ONE. 2016;11(4):e153274. https://doi.org/10.1371/journal.pone.0153274.

    Article  CAS  Google Scholar 

  93. Norenhag J, Du J, Olovsson M, et al. The vaginal microbiota, human papillomavirus and cervical dysplasia: a systematic review and network meta-analysis. BJOG. 2019;127(2):171–80. https://doi.org/10.1111/1471-0528.15854.

    Article  PubMed  Google Scholar 

  94. Brotman RM, Shardell MD, Gajer P, et al. Interplay between the temporal dynamics of the vaginal microbiota and human papillomavirus detection. J Infect Dis. 2014;210(11):1723–33. https://doi.org/10.1093/infdis/jiu330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Usyk M, Zolnik CP, Castle PE, et al. Cervicovaginal microbiome and natural history of HPV in a longitudinal study. PLoS Pathog. 2020;16(3):e1008376. https://doi.org/10.1371/journal.ppat.1008376.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Di Paola M, Sani C, Clemente AM, et al. Characterization of cervico-vaginal microbiota in women developing persistent high-risk human papillomavirus infection. Sci Rep. 2017;7(1):10200. https://doi.org/10.1038/s41598-017-09842-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Nilsen T, Swedek I, Lagenaur LA, et al. Novel selective inhibition of Lactobacillus iners by Lactobacillus-Derived Bacteriocins. Appl Environ Microbiol. 2020;86(20):e1520–94. https://doi.org/10.1128/AEM.01594-20.

    Article  Google Scholar 

  98. Sharifian K, Shoja Z, Jalilvand S. The interplay between human papillomavirus and vaginal microbiota in cervical cancer development. Virol J. 2023;20(1):73. https://doi.org/10.1186/s12985-023-02037-8.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Basavaprabhu HN, Sonu KS, Prabha R. Mechanistic insights into the action of probiotics against bacterial vaginosis and its mediated preterm birth: an overview. Microb Pathog. 2020;141:104029. https://doi.org/10.1016/j.micpath.2020.104029.

    Article  CAS  PubMed  Google Scholar 

  100. Witkin SS, Mendes-Soares H, Linhares IM, et al. Influence of vaginal bacteria and D- and L-lactic acid isomers on vaginal extracellular matrix metalloproteinase inducer: implications for protection against upper genital tract infections. mBio. 2013;4(4):e413–60. https://doi.org/10.1128/mBio.00460-13.

    Article  CAS  Google Scholar 

  101. Tachedjian G, Aldunate M, Bradshaw CS, et al. The role of lactic acid production by probiotic Lactobacillus species in vaginal health. Res Microbiol. 2017;168(9–10):782–92. https://doi.org/10.1016/j.resmic.2017.04.001.

    Article  CAS  PubMed  Google Scholar 

  102. Zheng N, Guo R, Wang J, et al. Contribution of Lactobacillus iners to Vaginal Health and diseases: a systematic review. Front Cell Infect Microbiol. 2021;11:792787. https://doi.org/10.3389/fcimb.2021.792787.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Beghini J, Linhares IM, Giraldo PC, et al. Differential expression of lactic acid isomers, extracellular matrix metalloproteinase inducer, and matrix metalloproteinase-8 in vaginal fluid from women with vaginal disorders. BJOG. 2015;122(12):1580–5. https://doi.org/10.1111/1471-0528.13072.

    Article  CAS  PubMed  Google Scholar 

  104. Ragaliauskas T, Plečkaitytė M, Jankunec M, et al. Inerolysin and vaginolysin, the cytolysins implicated in vaginal dysbiosis, differently impair molecular integrity of phospholipid membranes. Sci Rep. 2019;9(1):10606. https://doi.org/10.1038/s41598-019-47043-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Nowak RG, Randis TM, Desai P, et al. Higher levels of a cytotoxic protein, Vaginolysin, in Lactobacillus-Deficient Community State Types at the Vaginal Mucosa. Sex Transm Dis. 2018;45(4):e14–7. https://doi.org/10.1097/OLQ.0000000000000774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Colbert LE, El AM, Wang R, et al. Tumor-resident Lactobacillus iners confer chemoradiation resistance through lactate-induced metabolic rewiring. Cancer Cell. 2023;41(11):1945–62. https://doi.org/10.1016/j.ccell.2023.09.012.

    Article  CAS  PubMed  Google Scholar 

  107. Aldunate M, Srbinovski D, Hearps AC, et al. Antimicrobial and immune modulatory effects of lactic acid and short chain fatty acids produced by vaginal microbiota associated with eubiosis and bacterial vaginosis. Front Physiol. 2015;6:164. https://doi.org/10.3389/fphys.2015.00164.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Laniewski P, Herbst-Kralovetz MM. Bacterial vaginosis and health-associated bacteria modulate the immunometabolic landscape in 3D model of human cervix. NPJ Biofilms Microbiomes. 2021;7(1):88. https://doi.org/10.1038/s41522-021-00259-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Liu C, Li J, Shi W, et al. Progranulin regulates inflammation and tumor. Antiinflamm Antiallergy Agents Med Chem. 2020;19(2):88–102. https://doi.org/10.2174/1871523018666190724124214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Usyk M, Schlecht NF, Pickering S, et al. molBV reveals immune landscape of bacterial vaginosis and predicts human papillomavirus infection natural history. Nat Commun. 2022;13(1):233. https://doi.org/10.1038/s41467-021-27628-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ravel J, Moreno I, Simón C. Bacterial vaginosis and its association with infertility, endometritis, and pelvic inflammatory disease. Am J Obstet Gynecol. 2021;224(3):251–7. https://doi.org/10.1016/j.ajog.2020.10.019.

    Article  CAS  PubMed  Google Scholar 

  112. Lee JE, Lee S, Lee H, et al. Association of the vaginal microbiota with human papillomavirus infection in a Korean twin cohort. PLoS ONE. 2013;8(5):e63514. https://doi.org/10.1371/journal.pone.0063514.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Plisko O, Zodzika J, Jermakova I, et al. Aerobic vaginitis—underestimated risk factor for cervical intraepithelial neoplasia. Diagnostics (Basel). 2021;11(1):97. https://doi.org/10.3390/diagnostics11010097.

    Article  PubMed  Google Scholar 

  114. Wang C, Fan A, Li H, et al. Vaginal bacterial profiles of aerobic vaginitis: a case-control study. Diagn Microbiol Infect Dis. 2020;96(4):114981. https://doi.org/10.1016/j.diagmicrobio.2019.114981.

    Article  CAS  PubMed  Google Scholar 

  115. Ilhan ZE, Łaniewski P, Thomas N, et al. Deciphering the complex interplay between microbiota, HPV, inflammation and cancer through cervicovaginal metabolic profiling. EBioMedicine. 2019;44:675–90. https://doi.org/10.1016/j.ebiom.2019.04.028.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Borgogna JC, Shardell MD, Santori EK, et al. The vaginal metabolome and microbiota of cervical HPV-positive and HPV‐negative women: a cross‐sectional analysis. BJOG. 2019;127(2):182–92. https://doi.org/10.1111/1471-0528.15981.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Srinivasan S, Morgan MT, Fiedler TL, et al. Metabolic signatures of bacterial vaginosis. mBio. 2015;6(2):e204–15. https://doi.org/10.1128/mBio.00204-15.

    Article  CAS  Google Scholar 

  118. Liu L, Chen Y, Chen J, et al. Integrated metagenomics and metabolomics analysis of third-trimester pregnant women with premature membrane rupture: a pilot study. Ann Transl Med. 2021;9(23):1724. https://doi.org/10.21037/atm-21-5539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Nelson TM, Borgogna JL, Brotman RM, et al. Vaginal biogenic amines: biomarkers of bacterial vaginosis or precursors to vaginal dysbiosis? Front Physiol. 2015;6:253. https://doi.org/10.3389/fphys.2015.00253.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Borgogna JC, Shardell MD, Grace SG, et al. Biogenic amines increase the odds of bacterial vaginosis and affect the growth of and lactic acid production by vaginal Lactobacillus spp. Appl Environ Microbiol. 2021;87(10):e3020–68. https://doi.org/10.1128/AEM.03068-20.

    Article  Google Scholar 

  121. Fraszczak K, Barczynski B, Kondracka A. Does Lactobacillus exert a protective effect on the development of cervical and endometrial Cancer in women? Cancers (Basel). 2022;14(19):4909. https://doi.org/10.3390/cancers14194909.

    Article  CAS  PubMed  Google Scholar 

  122. Parada Venegas D, De la Fuente MK, Landskron G, et al. Short chain fatty acids (SCFAs)-Mediated gut epithelial and Immune Regulation and its relevance for inflammatory Bowel diseases. Front Immunol. 2019;10:277. https://doi.org/10.3389/fimmu.2019.00277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Delgado-Diaz DJ, Tyssen D, Hayward JA, et al. Distinct Immune responses elicited from Cervicovaginal Epithelial cells by Lactic Acid and short chain fatty acids Associated with Optimal and non-optimal vaginal microbiota. Front Cell Infect Microbiol. 2019;9:446. https://doi.org/10.3389/fcimb.2019.00446.

    Article  CAS  PubMed  Google Scholar 

  124. Mirzaei R, Kavyani B, Nabizadeh E, et al. Microbiota metabolites in the female reproductive system: focused on the short-chain fatty acids. Heliyon. 2023;9(3):e14562. https://doi.org/10.1016/j.heliyon.2023.e14562.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Torcia M. Interplay among vaginal Microbiome, Immune Response and sexually transmitted viral infections. Int J Mol Sci. 2019;20(2):266. https://doi.org/10.3390/ijms20020266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Garrett WS. Cancer and the microbiota. Science. 2015;348(6230):80–6. https://doi.org/10.1126/science.aaa4972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Galdiero MR, Marone G, Mantovani A. Cancer inflammation and cytokines. Cold Spring Harb Perspect Biol. 2018;10(8):a28662. https://doi.org/10.1101/cshperspect.a028662.

    Article  CAS  Google Scholar 

  128. Jayshree RS. The Immune Microenvironment in Human Papilloma Virus-Induced Cervical lesions-evidence for Estrogen as an Immunomodulator. Front Cell Infect Microbiol. 2021;11:649815. https://doi.org/10.3389/fcimb.2021.649815.

    Article  CAS  Google Scholar 

  129. Pavlova NN, Zhu J, Thompson CB. The hallmarks of cancer metabolism: still emerging. Cell Metab. 2022;34(3):355–77. https://doi.org/10.1016/j.cmet.2022.01.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Shannon B, Gajer P, Yi TJ, et al. Distinct effects of the cervicovaginal microbiota and Herpes Simplex Type 2 infection on female genital Tract Immunology. J Infect Dis. 2017;215(9):1366–75. https://doi.org/10.1093/infdis/jix088.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Hearps AC, Tyssen D, Srbinovski D, et al. Vaginal lactic acid elicits an anti-inflammatory response from human cervicovaginal epithelial cells and inhibits production of pro-inflammatory mediators associated with HIV acquisition. Mucosal Immunol. 2017;10(6):1480–90. https://doi.org/10.1038/mi.2017.27.

    Article  CAS  PubMed  Google Scholar 

  132. McKenzie R, Maarsingh JD, Łaniewski P, et al. Immunometabolic Analysis of Mobiluncus mulieris and Eggerthella sp. Reveals Novel insights into their pathogenic contributions to the hallmarks of bacterial vaginosis. Front Cell Infect Microbiol. 2021;11:759697. https://doi.org/10.3389/fcimb.2021.759697.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. van Teijlingen NH, Helgers LC, Zijlstra-Willems EM, et al. Vaginal dysbiosis associated-bacteria megasphaera elsdenii and Prevotella timonensis induce immune activation via dendritic cells. J Reprod Immunol. 2020;138:103085. https://doi.org/10.1016/j.jri.2020.103085.

    Article  CAS  PubMed  Google Scholar 

  134. Liu CW, Su BC, Chen JY. Tilapia Piscidin 4 (TP4) reprograms M1 macrophages to M2 phenotypes in cell models of Gardnerella vaginalis-Induced Vaginosis. Front Immunol. 2021;12:773013. https://doi.org/10.3389/fimmu.2021.773013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Lin F, Zhang B, Shi Q, et al. The conditioned medium of Lactobacillus rhamnoides GG regulates Microglia/Macrophage polarization and improves functional recovery after spinal cord Injury in rats. Biomed Res Int. 2021;2021:3376496. https://doi.org/10.1155/2021/3376496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Kudela E, Liskova A, Samec M, et al. The interplay between the vaginal microbiome and innate immunity in the focus of predictive, preventive, and personalized medical approach to combat HPV-induced cervical cancer. EPMA J. 2021;12(2):199–220. https://doi.org/10.1007/s13167-021-00244-3.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Arokiyaraj S, Seo SS, Kwon M, et al. Association of cervical microbial community with persistence, clearance and negativity of human papillomavirus in Korean women: a longitudinal study. Sci Rep. 2018;8(1):15479. https://doi.org/10.1038/s41598-018-33750-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Deruaz M, Luster AD. Chemokine-mediated immune responses in the female genital tract mucosa. Immunol Cell Biol. 2015;93(4):347–54. https://doi.org/10.1038/icb.2015.20.

    Article  CAS  PubMed  Google Scholar 

  139. Jang SE, Jeong JJ, Choi SY, et al. Lactobacillus rhamnosus HN001 and Lactobacillus acidophilus La-14 attenuate Gardnerella vaginalis-infected bacterial vaginosis in mice. Nutrients. 2017;9(6):531. https://doi.org/10.3390/nu9060531.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. De Seta F, Campisciano G, Zanotta N, et al. The Vaginal Community State types Microbiome-Immune Network as key factor for bacterial vaginosis and aerobic vaginitis. Front Microbiol. 2019;10:2451. https://doi.org/10.3389/fmicb.2019.02451.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Łaniewski P, Cui H, Roe DJ, et al. Features of the cervicovaginal microenvironment drive cancer biomarker signatures in patients across cervical carcinogenesis. Sci Rep. 2019;9(1):7333. https://doi.org/10.1038/s41598-019-43849-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Wei W, Xie LZ, Xia Q, et al. The role of vaginal microecology in the cervical cancer. J Obstet Gynaecol Res. 2022;48(9):2237–54. https://doi.org/10.1111/jog.15359.

    Article  CAS  PubMed  Google Scholar 

  143. Lebeau A, Bruyere D, Roncarati P, et al. HPV infection alters vaginal microbiome through down-regulating host mucosal innate peptides used by Lactobacilli as amino acid sources. Nat Commun. 2022;13(1):1076. https://doi.org/10.1038/s41467-022-28724-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Chen Y, Qiu X, Wang W, et al. Human papillomavirus infection and cervical intraepithelial neoplasia progression are associated with increased vaginal microbiome diversity in a Chinese cohort. BMC Infect Dis. 2020;20(1):629. https://doi.org/10.1186/s12879-020-05324-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Liu H, Liang H, Li D, et al. Association of Cervical Dysbacteriosis, HPV Oncogene expression, and cervical lesion progression. Microbiol Spectr. 2022;10(5):e15122. https://doi.org/10.1128/spectrum.00151-22.

    Article  CAS  Google Scholar 

  146. Mitra A, MacIntyre DA, Ntritsos G, et al. The vaginal microbiota associates with the regression of untreated cervical intraepithelial neoplasia 2 lesions. Nat Commun. 2020;11(1):1999. https://doi.org/10.1038/s41467-020-15856-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Mitra A, MacIntyre DA, Lee YS, et al. Cervical intraepithelial neoplasia disease progression is associated with increased vaginal microbiome diversity. Sci Rep. 2015;5(1):16865. https://doi.org/10.1038/srep16865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Kang G, Jung D, Lee YH, et al. Potential Association between vaginal microbiota and cervical carcinogenesis in Korean women: a Cohort Study. Microorganisms. 2021;9(2):294. https://doi.org/10.3390/microorganisms9020294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Wang Z, Xiao R, Huang J, et al. The diversity of Vaginal Microbiota predicts Neoadjuvant Chemotherapy responsiveness in locally Advanced Cervical Cancer. Microb Ecol. 2022;84(1):302–13. https://doi.org/10.1007/s00248-021-01800-0.

    Article  CAS  PubMed  Google Scholar 

  150. Zhang H, Lu J, Lu Y, et al. Cervical microbiome is altered in cervical intraepithelial neoplasia after loop electrosurgical excision procedure in China. Sci Rep. 2018;8(1):4923. https://doi.org/10.1038/s41598-018-23389-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Wiik J, Sengpiel V, Kyrgiou M, et al. Cervical microbiota in women with cervical intra-epithelial neoplasia, prior to and after local excisional treatment, a Norwegian cohort study. BMC Womens Health. 2019;19(1):30. https://doi.org/10.1186/s12905-019-0727-0.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Mitra A, MacIntyre DA, Paraskevaidi M, et al. The vaginal microbiota and innate immunity after local excisional treatment for cervical intraepithelial neoplasia. Genome Med. 2021;13(1):176. https://doi.org/10.1186/s13073-021-00977-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Bai J, Jhaney I, Daniel G, et al. Pilot Study of Vaginal Microbiome using QIIME 2™ in Women with Gynecologic Cancer before and after Radiation Therapy. Oncol Nurs Forum. 2019;46(2):E48–59. https://doi.org/10.1188/19.ONF.E48-E59.

    Article  PubMed  Google Scholar 

  154. Tsementzi D, Pena Gonzalez A, Bai J, et al. Comparison of vaginal microbiota in gynecologic cancer patients pre- and post‐radiation therapy and healthy women. Cancer Med. 2020;9(11):3714–24. https://doi.org/10.1002/cam4.3027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Morris L, Do V, Chard J, et al. Radiation-induced vaginal stenosis: current perspectives. Int J Womens Health. 2017;9:273–9. https://doi.org/10.2147/IJWH.S106796.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Chase D, Goulder A, Zenhausern F, et al. The vaginal and gastrointestinal microbiomes in gynecologic cancers: a review of applications in etiology, symptoms and treatment. Gynecol Oncol. 2015;138(1):190–200. https://doi.org/10.1016/j.ygyno.2015.04.036.

    Article  PubMed  Google Scholar 

  157. Chrysostomou D, Roberts LA, Marchesi JR, et al. Gut microbiota modulation of efficacy and toxicity of Cancer Chemotherapy and Immunotherapy. Gastroenterology. 2023;164(2):198–213. https://doi.org/10.1053/j.gastro.2022.10.018.

    Article  CAS  PubMed  Google Scholar 

  158. Santoni M, Piva F, Conti A, et al. Re: gut Microbiome influences Efficacy of PD-1-based Immunotherapy against epithelial tumors. Eur Urol. 2018;74(4):521–2. https://doi.org/10.1016/j.eururo.2018.05.033.

    Article  PubMed  Google Scholar 

  159. Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8. https://doi.org/10.1126/science.aao3290.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–9. https://doi.org/10.1126/science.aac4255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Wilkinson EM, Ilhan ZE, Herbst-Kralovetz MM. Microbiota–drug interactions: impact on metabolism and efficacy of therapeutics. Maturitas. 2018;112:53–63. https://doi.org/10.1016/j.maturitas.2018.03.012.

    Article  CAS  PubMed  Google Scholar 

  162. Wang M, Zhang L, Chang W, et al. The crosstalk between the gut microbiota and tumor immunity: implications for cancer progression and treatment outcomes. Front Immunol. 2023;13:1096551. https://doi.org/10.3389/fimmu.2022.1096551.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Chilakapati SR, Ricciuti J, Zsiros E. Microbiome and cancer immunotherapy. Curr Opin Biotechnol. 2020;65:114–7. https://doi.org/10.1016/j.copbio.2020.02.007.

    Article  CAS  PubMed  Google Scholar 

  164. Secombe KR, Coller JK, Gibson RJ, et al. The bidirectional interaction of the gut microbiome and the innate immune system: implications for chemotherapy-induced gastrointestinal toxicity. Int J Cancer. 2019;144(10):2365–76. https://doi.org/10.1002/ijc.31836.

    Article  CAS  PubMed  Google Scholar 

  165. Lozenov S, Krastev B, Nikolaev G, et al. Gut Microbiome Composition and its metabolites are a key regulating factor for Malignant Transformation, Metastasis and Antitumor Immunity. Int J Mol Sci. 2023;24(6):5978. https://doi.org/10.3390/ijms24065978.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Kamada N, Seo SU, Chen GY, et al. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013;13(5):321–35. https://doi.org/10.1038/nri3430.

    Article  CAS  PubMed  Google Scholar 

  167. Wong S, Slavcev RA. Treating cancer with infection: a review on bacterial cancer therapy. Lett Appl Microbiol. 2015;61(2):107–12. https://doi.org/10.1111/lam.12436.

    Article  CAS  PubMed  Google Scholar 

  168. Kobayashi O, Taguchi A, Nakajima T, et al. Immunotherapy that leverages HPV-specific immune responses for precancer lesions of cervical cancer. Taiwan J Obstet Gynecol. 2024;63(1):22–8. https://doi.org/10.1016/j.tjog.2023.10.002.

    Article  PubMed  Google Scholar 

  169. Recine N, Palma E, Domenici L, et al. Restoring vaginal microbiota: biological control of bacterial vaginosis. A prospective case–control study using Lactobacillus rhamnosus BMX 54 as adjuvant treatment against bacterial vaginosis. Arch Gynecol Obstet. 2016;293(1):101–7. https://doi.org/10.1007/s00404-015-3810-2.

    Article  PubMed  Google Scholar 

  170. De Alberti D, Russo R, Terruzzi F, et al. Lactobacilli vaginal colonisation after oral consumption of Respecta® complex: a randomised controlled pilot study. Arch Gynecol Obstet. 2015;292(4):861–7. https://doi.org/10.1007/s00404-015-3711-4.

    Article  PubMed  Google Scholar 

  171. Ansari A, Son D, Hur YM, et al. Lactobacillus Probiotics improve Vaginal Dysbiosis in Asymptomatic Women. Nutrients. 2023;15(8):1862. https://doi.org/10.3390/nu15081862.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Zeng M, Li X, Jiao X, et al. Roles of vaginal flora in human papillomavirus infection, virus persistence and clearance. Front Cell Infect Microbiol. 2023;12:1036869. https://doi.org/10.3389/fcimb.2022.1036869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Palma E, Recine N, Domenici L, et al. Long-term Lactobacillus rhamnosus BMX 54 application to restore a balanced vaginal ecosystem: a promising solution against HPV-infection. BMC Infect Dis. 2018;18(1):13. https://doi.org/10.1186/s12879-017-2938-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Lev-Sagie A, Goldman-Wohl D, Cohen Y, et al. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat Med. 2019;25(10):1500–4. https://doi.org/10.1038/s41591-019-0600-6.

    Article  CAS  PubMed  Google Scholar 

  175. Kumar L, Dwivedi M, Jain N, et al. The Female Reproductive Tract Microbiota: friends and Foe. Life (Basel). 2023;13(6):1313. https://doi.org/10.3390/life13061313.

    Article  CAS  PubMed  Google Scholar 

  176. Vaneechoutte M, Guschin A, Van Simaey L, et al. Emended description of Gardnerella vaginalis and description of Gardnerella leopoldii sp. nov., Gardnerella piotii sp. nov. and Gardnerella swidsinskii sp. nov., with delineation of 13 genomic species within the genus Gardnerella. Int J Syst Evol Microbiol. 2019;69(3):679–87. https://doi.org/10.1099/ijsem.0.003200.

    Article  CAS  PubMed  Google Scholar 

  177. Maarsingh JD, Łaniewski P, Herbst-Kralovetz MM. Immunometabolic and potential tumor-promoting changes in 3D cervical cell models infected with bacterial vaginosis-associated bacteria. Commun Biol. 2022;5(1):725. https://doi.org/10.1038/s42003-022-03681-6.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Ahrodia T, Das S, Bakshi S, et al. Structure, functions, and diversity of the healthy human microbiome. Prog Mol Biol Transl Sci. 2022;191(1):53–82. https://doi.org/10.1016/bs.pmbts.2022.07.003.

    Article  CAS  PubMed  Google Scholar 

  179. Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas. 2017;103:45–53. https://doi.org/10.1016/j.maturitas.2017.06.025.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This review is supported by Shanghai Changzheng Hospital from Doctoral Innovation Foundation for medical students in 8-year program (No. BSCX2022-32).

Author information

Authors and Affiliations

Authors

Contributions

J.S. wrote the main manuscript text, H.S. and J.C. were responsible for literature search and screening, X.G. prepared Figs. 1 and 2, and X.L. provided the conception and design of the review. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Xiaodi Gong or Xiaojun Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shen, J., Sun, H., Chu, J. et al. Cervicovaginal microbiota: a promising direction for prevention and treatment in cervical cancer. Infect Agents Cancer 19, 13 (2024). https://doi.org/10.1186/s13027-024-00573-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13027-024-00573-8

Keywords