Skip to main content
  • Systematic Review
  • Open access
  • Published:

Associations of the gut, cervical, and vaginal microbiota with cervical cancer: a systematic review and meta-analysis

Abstract

Background

An increasing number of studies indicate that the gut, cervical, and vaginal microbiota may play crucial roles in the development of cervical cancer (CC). However, the interactions between the microbiota and the host are yet unknown. To address this gap, a systematic review and meta-analysis were conducted to assess the microbiota alterations in a variety of body locations, including the gut and genital tract.

Methods

Electronic searches of PubMed, Embase, Web of Science, and the Cochrane Library were conducted to retrieve eligible papers published from January 1, 2014, to January 1, 2024 (PROSPERO: CRD42024554433). This study was restricted to English-language studies reporting on alpha diversity, beta diversity, and relative abundance, as well as on patients with CC whose microbiota had been analyzed via next-generation sequencing technologies. To assess the risk of bias (RoB), we utilized the Newcastle‒Ottawa Quality Assessment Scale (NOS) and the ROBINS-I tool. For the meta-analysis, we employed Review Manager 5.4.

Results

Thirty-six eligible studies were included in this review. The Chao1 index (SMD = 0.96, [95% CI: 0.71, 1.21], I2 = 0%) and the Shannon index (SMD = 1.02, [95% CI: 0.53, 1.50], I2 = 85%) values from vaginal samples were significantly greater in patients than in the controls. In the cervical samples, the Shannon index value (SMD = 1.29, [95% CI: 0.61, 1.97], I2 = 93%) significantly increased, whereas the Chao1 index value did not significantly differ (SMD = 0.50, [95% CI: -0.46, 1.46], I2 = 89%). The Shannon index value (SMD = 0.25, [95% CI: -0.22, 0.72], I2 = 38%) did not significantly differ across the gut samples. The majority of studies (19/25) indicated that the patients and noncancer controls differed significantly in terms of beta diversity. Cancer-associated changes were observed, with a dramatic decrease in the Lactobacillus genus and significant increases in pathogenic bacteria, including the Anaerococcus, Peptostreptococcus, Porphyromonas, Prevotella, and Sneathia genera. Additionally, the impact of antineoplastic therapies on microbial diversity was inconsistently reported across several studies.

Conclusion

This systematic review elucidates the microbiota alterations associated with the prevalence of CC and its response to anti-tumor therapies, aiming to provide insights for future research directions and precision medicine strategies to enhance women’s quality of life.

Prospero registration

CRD42024554433.

Peer Review reports

Background

Cervical cancer (CC) is the fourth most common malignancy in women worldwide, following breast cancer, lung cancer, and colorectal cancer [1]. Globally, there were estimated to be 342,000 deaths and 604,000 new cases in 2020, with considerably higher rates in transitioning countries than in transitioned countries [1]. CC is caused primarily by persistent high-risk human papillomavirus (HPV) infection [2]. Despite the promotion of the HPV vaccine, early screening, and standardized treatment of CC involving surgery, chemotherapy, radiotherapy, and immunotherapy, the outcomes of CC treatment are still not satisfactory [3]. In addition to HPV infection, numerous other factors contribute to CC progression. With rapid advancements in sequencing technologies, an increasing number of studies have explored the role of the human microbiota in carcinogenesis [4].

The human microbiota is defined as the various communities of beneficial and pathogenic microorganisms, including bacteria, archaea, fungi, protists, and viruses, that exist within the unique environment of the human body [5]. Owing to its anatomical location, the cervix is tightly connected to the genital and gut microbiota [6]. Anatomically, the female reproductive system consists of an upper section (uterus, fallopian canals, and ovaries) and a lower portion (cervix and vagina) with distinct microenvironmental compositions [7]. The vaginal microbiota comprises the majority of microorganisms in the female reproductive tract and is divided into five categories on the basis of the dominant species of the Lactobacillus genus: Lactobacillus crispatus, Lactobacillus gasseri, Lactobacillus iners, anaerobic bacteria, and Lactobacillus jensenii [8]. Emerging evidence shows that the microbiota in the cervicovaginal tract plays a crucial role in the progression of CC [9,10,11].

The gut microbiota contains approximately 1000 species of microorganisms and is termed the “second genome” of the human body [12]. Emerging evidence indicates that the microorganisms in the gastrointestinal tract contribute to the development of cancers, such as gastric cancer, colorectal cancer, lung cancer, breast cancer, and CC [13,14,15,16,17,18]. The pathophysiological mechanisms of microbial carcinogenesis may involve failure of the epithelial barrier, chronic inflammation, immune dysregulation, and/or genotoxicity [19,20,21]. Moreover, research on preclinical mouse models and clinical cancer patients has demonstrated that gut microbial diversity and composition can influence antitumor immunity and affect the efficacy of immunotherapy in patients with melanoma and lung and kidney cancers [22].

Systematic reviews have focused on alterations in vaginal samples from patients with CC, indicating that Lactobacillus iners is a protective factor against carcinogenesis [23, 24]. However, further exploration is needed to focus simultaneously on microbiota alterations in both the genital tract and gut of CC patients. Therefore, we aimed to investigate the interaction between CC and the microbiota in the genital and gastrointestinal tracts and to evaluate treatment‒microbiota interactions. Here, we present a systematic review and meta-analysis to characterize the composition of the microbiota in CC patients compared with that in non-CC controls and to explore the impact of treatment on the microbiota.

Methods

The systematic review was preregistered with PROSPERO (CRD42024554433) and was conducted on the basis of the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines [25].

Search strategy and information sources

Literature searches for inclusion in this systematic review were conducted in PubMed, Embase, Web of Science, and the Cochrane Library, as described in Appendix S1. The search was restricted to studies involving adults with a diagnosis of CC and to studies published in the English language within the last decade. Studies including only nonhuman subjects were excluded. We also manually examined the reference lists of the retrieved publications to identify additional appropriate studies that were missed during the primary search.

Eligibility criteria and study selection

An initial screening for compliance was carried out by two independent reviewers who read the titles and abstracts of the literature before moving on to a full-text reading screen. After careful reading of any literature that met the inclusion requirements, data extraction was performed. The following inclusion criteria were used: (1) the target population included adult patients with CC; (2) sequencing was performed via high-throughput sequencing and assessment of α-diversity, β-diversity, and relative abundance; and (3) a control group was included for the following comparisons: comparing nontumor patients to assess the changes in the microbiota with or without cancer or comparing patients before and after antineoplastic treatment to assess the effects of such treatment on the microbiota; or (4) there were subgroups with different microbiota characteristics used to assess the effects of microbiota on treatment measures. Consensus and group discussion were the main methods used to resolve disagreements.

Data extraction and assessment of RoB

Two authors employed a preestablished template to extract data and then cross-checked for the following: authors, published year, nation, number of participants in each group, age, ethnicity, HPV status, smoking or drinking, sequencing technology, 16 S rRNA variable region, and sequencing platform, among others. The main areas of focus of this analysis were the microbiota composition at the community level (α and β diversity) and taxonomic discoveries at the phylum, family and genus levels (relative abundance).

We performed a quality assessment of the case‒control studies and nonrandomized trials on the basis of the Newcastle–Ottawa Scale (NOS) [26] and ROBINS-I stool [27], respectively. A score of 0 was assigned for a “no” or “unclear” answer, whereas a score of 1 was assigned for a “yes” answer. The NOS is scored on a scale of 9, with studies receiving a score of 6 or higher considered to be of good quality [28, 29]. Therefore, studies with an NOS score between 6 and 9 were included in our meta-analysis to enhance the reliability of the findings. Two researchers separately assessed the RoB, and when there was dispute, it was settled by consensus and group discussion.

Data synthesis

Data were extracted from text, tables, and figures for analysis. For those studies lacking original data, the WebPlotDigitizer online tool was used to extract and estimate data from the box plot as previously described [30,31,32]. Means (M) and standard deviations (SD) were calculated via a web tool (http://www.math.hkbu.edu.hk/~tongt/papers/median2mean.html) by converting medians, interquartile ranges, minimum values, and maximum values [33]. The meta-analysis employed bacterial diversity indices, a random effects model, and standardized mean difference (SMD) calculations to generate forest plots depicting microbiome variations between patients and controls. The I2 statistic was used to assess heterogeneity levels, which were categorized as low (25%), moderate (50%), or high (75%), and funnel plots were used to detect publication bias. Subgroup analyses were conducted on different samples. Review Manager version 5.4 was utilized for all the statistical analyses to synthesize the data and evaluate the outcomes.

Results

Study selection

The search yielded 1613 published articles from PubMed, Embase, Web of Science, and the Cochrane Library. After 456 duplicate records were removed, 1157 articles were screened for exclusion on the basis of non-English language or inappropriateness assessed after the titles and abstracts were read. In total, this systematic review and meta-analysis involved a detailed evaluation of 113 full-text articles, with 36 studies ultimately included (Fig. 1). These included 31 case‒control studies and 9 nonrandomized intervention studies, with four studies included in both the case‒control and nonrandomized intervention categories.

Fig. 1
figure 1

Flow Diagram of the Study Selection in the Systematic Literature Review

Study characteristics

The included studies spanned from 2014 to 2024 and were conducted across 12 countries, predominantly (21 [58.33%]) in East Asia (China, Korea), 12 (33.33%) in westernized populations (USA, Mexico, UK, Germany, Russia, Poland, Romania), and 3 (8.33%) in Africa (Nigeria, Ethiopia, Botswana) (Table S1). In total, 3333 participants were included in the studies; these participants included 929 patients, 1179 cervical intraepithelial neoplasia (CIN) controls, and 1225 healthy controls (HCs), with sample sizes ranging from 4 to 124. The participants’ average age was 43.39 years, with a standard deviation of 11.46 years. In addition, the mean BMI was 24.32, with a standard deviation of 4.46. The participants were divided into three groups according to their HPV infection status: HPV16/18, other HPVs, and uninfected, with 89.6% of the CC patients being infected with HPV. (Table S2).

Various methodologies were adopted for analyzing the composition and sequencing of samples (Table S3). Thirty-three studies employed 16 S rRNA sequencing techniques on the Illumina platform, whereas four studies employed shotgun metagenomic sequencing. The hypervariable region V3-V4 was the most frequently sequenced region (14/33), followed by region V4 alone (12/33) and other regions (7/33). The Illumina MiSeq, Illumina HiSeq, IonPGM and 454 GS platforms were primarily used for these sequencing analyses. The SILVA database (13/36) and the Greengenes database (8/36) were the most widely used databases.

Assessment of RoB

The RoB was determined on the basis of the type of research. The NOS scale was employed to evaluate the case‒control studies (Figure S1), with scores ranging from 6 to 8 (mean = 7.39). The nonresponse rate score was set to zero for all the studies since it was not described in the text. The ROBINS-I stool was utilized to assess the RoB in nonrandomized trials (Figure S2), with each study graded as low risk. No articles were excluded from this systematic review due to RoB.

Alpha diversity

The alpha diversity of microbiota communities was assessed in 27 of 31 case‒control studies and 8 of 9 nonrandomized studies, all of which provided precise data or statistical plots. The most commonly used index was the Shannon index, followed by the Simpson index, Chao1, and observed species. None of the funnel plots showed evidence of publication bias upon visual inspection (Figure S3).

On the basis of an analysis of studies comparing genital microbiome richness among CC patients, HCs, and CIN controls, ten studies revealed Chao1 indices in patients (n = 225), HCs (n = 474) and CIN controls (n = 356) (Figs. 2A and 3A) [18, 34,35,36,37,38,39,40,41,42]. The microbiota of the vaginal samples tended to significantly increase (HC control: SMD = 0.96 [95% CI: 0.71, 1.21], I2 = 0%; CIN control: SMD = 0.81 [95% CI: 0.55, 1.08], I2 = 0%), whereas in the cervical samples, no significantly different results were observed (HC control: SMD = 1.08 [95% CI: -0.28, 2.44], I2 = 95%; CIN control: SMD = 0.12 [95% CI: -0.16, 0.39], I2 = 0%). The observed species indices were reported in nine studies with patients (n = 211), HCs (n = 408) and CIN controls (n = 480) (Figs. 2B and 3B) [10, 37,38,39, 41,42,43,44,45]. The cervical sample indices did not significantly differ between patients and HCs (SMD = 0.50, [95% CI: -0.46, 1.46], I2 = 89%) or between patients and CIN controls (SMD = 0.15, [95% CI: -0.12, 0.43], I2 = 2%). In contrast, a notable increase was observed in the vaginal sample comparisons (HC control: SMD = 0.93, [95% CI: 0.61, 1.25], I2 = 30%; CIN control: SMD = 0.92, [95% CI: 0.52, 1.32], I2 = 52%).

Regarding the assessment of genital microbiome diversity, the Shannon index value was reported in 26 studies involving patients (n = 516), HCs (n = 996), and CIN controls (Figs. 2C and 3C) (n = 940) [10, 11, 18, 34, 36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57]. Both comparisons revealed a significant increase in cervical samples (HC control: SMD = 1.29 [95% CI: 0.61, 1.97], I2 = 93%; CIN control: SMD = 0.82 [95% CI: 0.19, 1.45], I2 = 91%) and vaginal samples (HC control: SMD = 1.02 [95% CI: 0.53, 1.50], I2 = 85%; CIN control: SMD = 0.84 [95% CI: 0.55, 1.13], I2 = 52%). The Simpson index value was reported in 14 studies involving patients (n = 337), HCs (n = 504), and CIN controls (n = 540) (Figs. 2D and 3D) [10, 18, 37, 39, 41, 42, 44, 47,48,49, 53,54,55,56]. Both comparisons revealed no significant difference in the cervical samples (HC control: SMD = 0.29 [95% CI: -0.32, 0.90], I2 = 86%; CIN control: SMD = 0.27 [95% CI: -0.29, 0.82], I2 = 82%), whereas the indices for the vaginal samples presented different results (HC control: SMD = -0.08 [95% CI: -0.84, 0.69], I2 = 90%; CIN control: SMD = 0.41 [95% CI: 0.01, 0.82], I2 = 58%).

Fig. 2
figure 2

Forest Plots of Alpha Diversity in the Microbiota of Patients with CC Compared with HC. (A) Chao1 (B) Observed species (C) Shannon index (D) Simpson index

Fig. 3
figure 3

Forest Plots of Alpha Diversity in the Microbiota of Patients with CC Compared with Patients with CIN. (A) Chao1 (B) Observed species (C) Shannon index (D) Simpson index

With respect to the gut microbiota, only three studies evaluated the microbial alterations between patients (n = 67) and HCs (n = 80) (Fig. 2). The Shannon index value was reported in 3 studies (SMD = 0.25, [95% CI: -0.22, 0.72], I2 = 38%]), the Chao1 index value was reported in 2 studies (SMD = 0.69, [95% CI: 0.14, 1.23], I2 = 0%]), the number of observed species was reported in 1 study (SMD = -0.72, [95% CI: -1.33, -0.10]), and the Simpson index value was reported in 1 study (SMD = 0.42, [95% CI: -0.71, 1.56]).

To better understand the heterogeneity among studies, we conducted a subgroup analysis based on geographic region, age group, sample collection techniques, and sequencing methods (Table S4). In the subgroup of participants aged ≤ 50 years, heterogeneity significantly decreased, as indicated by the I² value dropping to 0, suggesting that age variations likely contributed to the observed heterogeneity. However, heterogeneity remained high in the swab sampling group, the studies from China group, and the 16 S rRNA sequencing group. A downward trend was noted in these groups, indicating that these factors may influence the results. Further investigation is warranted to confirm these findings.

In nonrandomized trials, the impact of treatments on the microbiome has been the subject of attention. Data on the Shannon index in patients (posttreatment, n = 126) compared with controls (pretreatment, n = 169) were obtained from six studies [37, 40, 43, 58,59,60]. A significant decrease in posttreatment patients was shown by the pooled estimate (SMD = -0.43, [95% CI: -0.85, -0.02], I2 = 63%) (Fig. 4A). Only three publications reported the Chao1 estimator [33, 35, 38] (Fig. 4B), observed species [35, 41, 59] (Fig. 4C), and Simpson index [37, 60, 61] (Fig. 4D), and none of these values demonstrated statistically significant differences.

Fig. 4
figure 4

Forest Plots of Alpha Diversity in the Microbiota of Post-treatment Patients Compared with Pre-treatment Patients. (A) Shannon index (B) Chao1 (C) Observed species (D) Simpson index

Beta diversity

The microbial community compositions of the patient and control samples were compared using beta diversity. Twenty-five of the 36 studies reported beta diversity using various measures, with two studies comparing patients to controls as well as comparing the pre- to posttreatment conditions (Table S4). In 19 studies, significant differences in beta diversity were detected in 11 cases involving cervical samples, 4 cases involving vaginal samples, and 4 cases involving fecal samples compared with noncancer controls. However, four studies reported that the cancer patients did not differ significantly from the noncancer controls. While Łaniewski et al. [9] found no differences between cancer patients and controls, they did observe significant differences when participants were clustered on the basis of vaginal PH. Four studies compared the microbial communities of patients before and after antineoplastic therapy. Among them, two studies reported significant differences before and after radiation treatment, whereas the other two reported no significant differences.

Differential abundance of microbial taxa

All the investigations evaluated the relative abundance of microorganisms, and 23 of them reported significant differences between the patient and control groups [9, 18, 34,35,36, 38, 39, 41,42,43,44,45, 47,48,49, 51,52,53,54,55,56,57, 62]. Overall, seven phyla, 12 families, and 40 genera were identified from four samples: cervical sample, cervicovaginal sample, vaginal sample, and fecal sample.

The within-disorder comparison is illustrated below, with inconsistent findings categorized as “not consistent” (Fig. 5). The majority of consistently observed changes within disorders were replicated by only two studies, indicating a need for further investigation.

Fig. 5
figure 5

Summary of Changes in the Relative Abundance of Microbial Taxa from a Sample Category

Cancer-associated alterations

Cancer specificity was defined as consistent alterations observed in the same direction across three or more studies. Our findings revealed several cancer-associated alterations, including enrichment of the genera Anaerococcus, Peptostreptococcus, Porphyromonas, Prevotella, and Sneathia as well as depletion of the Lactobacillus genus (Figure S4). However, there is limited evidence for consistent enrichment of pathogenic bacteria such as Anaerococcus, Peptostreptococcus, Porphyromonas, Prevotella, and Sneathia, as the findings were not consistent across more than three studies. Regarding the Lactobacillus genus, CST I and CST III, dominated respectively by Lactobacillus crispatus and Lactobacillus iners, were positively associated with healthy microbiota, as demonstrated in seven and six publications, respectively. Overall, 18 out of 23 studies reported a significant decrease in Lactobacillus species in the cervical, cervicovaginal, and vaginal samples of CC patients, providing robust evidence of cancer specificity.

Treatment-associated alterations

In this meta-analysis, none of the patients in the 28 studies used either antibiotics or probiotics before sample collection. A total of 9 studies provided data on how antineoplastic treatment influences the relative abundance of the microbiota [35, 37, 40, 43, 58,59,60,61, 63], including 6 studies on chemoradiotherapy (CRT), 1 on pelvic radiotherapy, 1 on radical hysterectomy and 1 encompassing various antineoplastic treatments. Among these, only six studies assessed changes in the microbiota before and after therapy. At the genus level, one study reported a significant increase in the genera Varibaculum, Bosea, Actinotignum, and Propionimicrobium and a decrease in Cutibacterium, Acinetobacter, Rhodococcus, Amaricoccus, and Paracoccus in patients receiving antineoplastic treatments. Additionally, there was evidence of enrichment of Desulfovibrio and Turicibacter in patients undergoing radical hysterectomy and depletion of the genera Atopobium and Aerococcus in patients with CRT. However, these treatment-associated alterations have been reported in only single studies, indicating limited evidence and the need for further investigation.

Discussion

This systematic review included data on the cervical, vaginal, or gut microbiota in patients with CC. The present meta-analysis evaluated microbiological alterations by comparing CC patients with nontumor controls and examined changes before and after antineoplastic therapy. The findings are summarized as follows: [1] There was a slight increase in the effect size of the Chao1 and Shannon indices comparing CC with CIN; a moderate increase in this effect size in patients versus HCs; and a slight increase in the Shannon index value was compared between patients before and after treatment; [2] significant differences in beta diversity were consistently observed in the majority of studies (19/25); [3] cancer-associated alterations included a significant decrease in Lactobacillus species in all samples, identified in the majority of studies (18/23); and [4] treatment-associated alterations were noted, but evidence was limited and requires further verification.

With respect to alpha diversity (within samples), specific indices, such as the Chao1 estimator for richness, the Simpson index, and the Shannon index for community diversity (both richness and evenness), were used to assess the diversity and abundance of the bacterial microbiota. In the vaginal microbiota, both richness (Chao1 and observed species) and diversity (Shannon index) significantly increased in CC patients compared with HCs or CIN patients. Furthermore, in the cervical microbiota, all richness and diversity indices showed no differences except for a significant increase in the Shannon index value in CC patients compared with HCs. Overall, greater microbiota diversity in the genital tract was observed in more severe pathological conditions, suggesting that microbial dysbiosis of the genital tract, typified by the replacement of Lactobacillus species with diverse anaerobic bacteria, may be a contributing factor to CC progression [6, 10]. Previous studies have demonstrated that some intestinal bacteria in colorectal cancer can promote carcinogenesis by modulating inflammatory factors such as NF-κB and IL-6 [19, 64]. Moreover, several cross-sectional studies have shown correlations between IL-4 and Fusobacterium spp. [11], as well as between proinflammatory (IL-36γ) and non-Lactobacillus dominance [9]. Thus, chronic inflammation may be one of the possible mechanisms by which bacteria associated with CC contribute to cancer progression.

The gut microbiome interacts with the reproductive tract microbiota through direct or estrogen-mediated mechanisms. The proximity of the vagina and anus results in a shared microbiota, with over 30% of bacterial species in common, including Firmicutes and Bacteroidetes [65]. The gut microbiota may partially function as a reservoir for the vagina microbiota. Furthermore, gut microbial diversity has been shown to influence the composition of vaginal microbiota through the estrogen-mediated gut‒vaginal axis. This process is primarily mediated by intestinal bacteria capable of metabolizing estrogen and promoting the growth of Lactobacillus spp. through estrogen degradation and reabsorption [66,67,68,69]. Microbe-secreted β-glucuronidase and β-glucosidase contribute to the deconjugation of estrogens, thereby facilitating their reabsorption into the circulation. In the female reproductive tract, free estrogen binds to receptors and transmits intracellular signals that increase gluconeogenesis and induce other physiological changes, such as mucus production and epithelial thickening. Enhanced glycogen synthesis facilitates the proliferation of lactobacilli, which are essential for maintaining vaginal homeostasis [67, 69].

In addition to linking with the gut-vaginal and gut-brain axes, the gut microbiota also impacts systemic diseases, including cancer, through its physiological activities in the gastrointestinal system, which regulates immune function and metabolic processes. Alterations in gut microbiota significantly influence disease through the production of metabolites such as short-chain fatty acids (SCFA), tryptophan (Trp), and bile acid (BA) metabolites, which are synthesized by diverse microorganisms [70, 71]. These metabolites can promote the differentiation and function of immunosuppressive cells, such as regulatory T cells, while inhibiting inflammatory cells, thereby contributing to the maintenance of both intestinal and systemic immune homeostasis [72, 73]. Additionally, dysbiosis of the gut microbiota can compromise the intestinal barrier and enhance lipopolysaccharide-induced inflammation, allowing harmful molecules from gut microorganisms to enter the bloodstream through capillaries, thus contributing to the development of distant diseases [74, 75]. However, this review highlights contentious conclusions regarding diversity indices in a small number of studies, showing no significant variations in the Shannon and Simpson indices, a decline in observed species, and an increase in Chao1, thus calling for further research to explore the potential role of gut microbiome in the progression of cervical cancer.

Beta diversity measures are widely employed to assess the similarity between communities, with the majority of studies demonstrating significant differences between patients and controls. Although some studies have shown no significant difference, beta diversity may still serve as a promising diagnostic biomarker.

Although there is limited evidence demonstrating differences between patients before and after treatment in terms of treatment-associated alterations, available studies suggest that antitumor therapy may negatively impact the microbiota composition. However, the treatment-related alterations in microbial communities exhibited inconsistencies across studies in our meta-analysis. Differences in treatment regimens may substantially contribute to this variability. Prior studies indicated that therapies for CC, such as radiotherapy, chemotherapy, and immunotherapy, can compromise the intestinal epithelial barrier, thus facilitating the proliferation of opportunistic pathogenic bacteria [76,77,78]. Furthermore, radiotherapy can directly influence the microbiota in the gut or vagina, particularly in areas exposed, such as the cervix [79]. Chemotherapy, owing to its systemic toxic effects, leads to immunosuppression and modifies various metabolic pathways, causing significant dysbiosis and the selective enhancement of specific drug-resistant pathogens [80,81,82]. Immunotherapy might affect the composition of the microbiota by altering microbe-related signaling or metabolic functions controlled by host immunity [83]. Combination therapy, such as concurrent chemoradiotherapy, may affect the microbiota through intricate mechanisms, leading to diverse changes in microbial composition. Additionally, microbial diversity typically diminishes with the progression of antitumor therapy [63]. The variability in microbial sampling time points among the studies analyzed may have contributed to the observed inconsistencies. Dysbiosis during antineoplastic treatments may serve as a mechanism potentially influencing both the response and toxicity to therapies [84, 85].

Additionally, our results suggest that CC may lead to cancer-associated alterations in the microbiota. Consistently, the abundance of the Lactobacillus species decreased markedly with the progression of cervical lesions, whereas the abundances of pathogenic bacteria, including Anaerococcus, Peptostreptococcus, Porphyromonas, Prevotella, and Sneathia, increased significantly. Notably, the Lactobacillus species were found to be favorably associated with a healthy state, preventing pathogen invasion in the microenvironment by inhibiting pathogen adherence and producing antimicrobial agents, among other mechanisms [66, 86, 87]. A reduction in lactobacilli can disturb the balance of the vaginal ecosystem, promoting the overgrowth of pathogenic bacteria. Specific species linked to a high diversity of microbiome composition, such as Prevotella, bacterial vaginosis-associated bacteria (BVAB), and Sneathia, are capable of producing sialidases, resulting in mucus breakdown and rendering the cervical epithelium susceptible to tissue damage [88]. BVAB, including Gardnerella vaginalis, exhibits proteomic characteristics associated with immune activation, apoptosis, epithelial integrity disruption, and impaired wound healing [89]. Conversely, pathogenic bacteria trigger chronic inflammation and secrete bacterial genotoxins, which contribute to the persistence of inflammation and establish conditions conducive to cancer progression, including cellular and DNA damage [90, 91]. Tissue damage and the establishment of an inflammatory environment increase the susceptibility of tissues to HPV infection [92]. HPV persistence is a known causative factor of CC through the integration of viral nucleic acid into host genomes and the induction of mutations in host cells [19, 93, 94]. Previous studies have shown that bacteria such as Prevotella and Sneathia are linked to persistent HPV infection [88, 95,96,97], although the precise mechanism of interaction between the two remains unclear and warrants further investigation.

Accumulating evidence indicates that microbial dysbiosis is linked to cancer progression, as well as the efficacy and side effects of anti-cancer treatments, suggesting that microbial-targeted therapy holds significant clinical promise. Microbial dysbiosis is associated with persistent HPV infection and the advancement of CIN during tumor progression [88]. However, the diverse proinflammatory environment in women with CIN cannot be entirely rectified through surgical resection or HPV clearance. The inability to restore the Lactobacillus-rich CST may elucidate the persistently elevated risk of recurrence of pre-invasive and invasive disease in women [98]. The vaginal probiotic Lactobacillus, specifically the L. crispatus strain CTV-05 (administered as a vaginal suppository, LACTIN-V), is primarily utilized for treating bacterial vaginosis and is currently in clinical trials, indicating the potential of vaginal probiotics in modulating the vaginal microbiome [99, 100]. The microbiome can simultaneously influence the efficacy of anti-cancer therapies through various mechanisms, including translocation, immune modulation, metabolism, and enzymatic degradation [101, 102]. The composition of the gut microbiota affects the efficacy of chemotherapeutic agents, such as irinotecan, by modifying drug metabolism and modulating the host immune response [103]. This modulation also influences radiotherapy and immunotherapy, including PD-1/PD-L1 blockade, primarily by mediating T cell activation, enhancing T cell priming, and facilitating T cell accumulation at tumor sites [22, 104,105,106]. Utilizing antibiotics or other inhibitors to reduce gut microbiota, supplementation through fecal microbiota transplantation (FMT) or vaginal microbiota transplantation (VMT), and enhancement with prebiotics or probiotics may serve to prevent or alleviate cervical cancer and improve responses to cancer therapy.

Overall, the relationship between the microbiota (cervical, vaginal and gut) and CC, as well as its relationship with treatment, is relatively poorly understood. Dysbiosis may affect the development of CC through mechanisms such as chronic inflammation and persistent HPV infection. Local dysbiosis caused by antitumor therapies may influence treatment side effects. However, these mechanisms are still unclear and need to be explored in more appropriate mechanistic studies.

This meta-analysis has several limitations. First, the majority of studies contained modest sample sizes, which indicates that our analysis may be preliminary and that larger sample sizes are necessary. Additionally, inadequate data prevented the inclusion of variables such as the Pielou index, Good’s coverage, and the ACE index in this meta-analysis. Second, our study utilized multiple sequencing technologies, specifically 16 S rRNA gene sequencing, 16 S rDNA gene sequencing, and shotgun metagenomic sequencing [107,108,109]. Although 16 S rRNA gene sequencing serves as the standard approach for microbial community analysis and yields important bacterial taxonomic data, it predominantly depends on the amplification of specific regions of the 16 S rRNA gene, which may restrict its resolution in distinguishing certain species or subspecies [107]. Shotgun metagenomic sequencing provides a more comprehensive analysis of microbial communities and offers functional insights into microorganisms [109]. However, it exhibits greater vulnerability to sample contamination, biases, and difficulties in data processing. The heterogeneity among studies resulting from varying sequencing technologies may have introduced bias into our results. Moreover, differences in technical platforms result in variations in sequencing depth, coverage, and error rates, which can impact microbiota diversity estimation and lead to discrepancies in species classification, potentially affecting the analysis outcomes [110].

Third, this systematic review included several nonrandomized trial studies, offering preliminary insights into the impact of antineoplastic treatments on the microbiota. However, these studies were limited in number and exhibited inconsistencies in their findings, thus providing insufficient evidence to draw firm conclusions on how anticancer treatments affect the microbiota, thereby necessitating additional research. Finally, we did not emphasize the functional impact of the microbiota on the progression of cancer, although previous studies have demonstrated associations between the microbiota (e.g., Fusobacterium spp., non-Lactobacillus dominance) and mediators such as proinflammatory (IL-36γ), chemotactic (MIP1β), hematopoietic (FLT3 ligand) and adaptive immune response cytokines (IL-2, IL-4 and soluble CD40 ligand) [9, 11]. This finding emphasizes how vital functional analysis is a crucial tool for comprehending how host‒microbiome interactions contribute to CC.

Conclusion

In conclusion, our study highlights distinct changes in the genital and gut microbiota among patients with CC. These alterations are characterized by a notable increase in alpha diversity, a substantial decrease in the relative abundance of Lactobacillus spp. and an increase in the relative abundance of pathogenic microbes compared with those of the controls. Furthermore, treatment-associated microbiota alterations have also been incipient, which may provide a cornerstone for the interaction of antitumor therapies with the microbiota. Therefore, more clinical studies are warranted to investigate treatment‒microbiome interactions and host‒microbiome interactions in the context of CC and in vivo and in vitro experiments to explore the mechanism behind this phenomenon. Finally, a clearer understanding of the role of microorganisms in the progression of CC and their interactions with anticancer therapies may lead to new opportunities for cancer prevention, precision therapy, and improved quality of life for women.

Data availability

The data that support the findings of this study are available from the corresponding author, [MHC], upon reasonable request.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, 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.

    Article  PubMed  Google Scholar 

  2. Crosbie EJ, Einstein MH, Franceschi S, Kitchener HC. Human papillomavirus and cervical cancer. Lancet. 2013;382(9895):889–99.

    Article  PubMed  Google Scholar 

  3. Meijer CJ, Snijders PJ. Cervical cancer in 2013: screening comes of age and treatment progress continues. Nat Rev Clin Oncol. 2014;11(2):77–8.

    Article  CAS  PubMed  Google Scholar 

  4. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med. 2019;25(3):377–88.

    Article  CAS  PubMed  Google Scholar 

  5. Marchesi JR, Ravel J. The vocabulary of microbiome research: a proposal. Microbiome. 2015;3:31.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Laniewski P, Ilhan ZE, Herbst-Kralovetz MM. The microbiome and gynaecological cancer development, prevention and therapy. Nat Rev Urol. 2020;17(4):232–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A. 2011;108(1):4680–7.

    Article  CAS  PubMed  Google Scholar 

  8. Gajer P, Brotman RM, Bai G, Sakamoto J, Schütte UM, Zhong X, et al. Temporal dynamics of the human vaginal microbiota. Sci Transl Med. 2012;4(132):132ra52.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Łaniewski P, Barnes D, Goulder A, Cui H, Roe DJ, Chase DM, 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.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mitra A, MacIntyre DA, Lee YS, Smith A, Marchesi JR, Lehne B, et al. Cervical intraepithelial neoplasia disease progression is associated with increased vaginal microbiome diversity. Sci Rep. 2015;5:16865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Audirac-Chalifour A, Torres-Poveda K, Bahena-Román M, Téllez-Sosa J, Martínez-Barnetche J, Cortina-Ceballos B, et al. Cervical Microbiome and Cytokine Profile at various stages of cervical Cancer: a pilot study. PLoS ONE. 2016;11(4):e0153274.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Zhu B, Wang X, Li L. Human gut microbiome: the second genome of human body. Protein Cell. 2010;1(8):718–25.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021;371(6536):eabc4552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, et al. Fusobacterium nucleatum promotes Chemoresistance to Colorectal Cancer by modulating Autophagy. Cell. 2017;170(3):548–e6316.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Knippel RJ, Drewes JL, Sears CL. The Cancer Microbiome: recent highlights and knowledge gaps. Cancer Discov. 2021;11(10):2378–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, et al. Commensal microbiota promote Lung Cancer Development via γδ T cells. Cell. 2019;176(5):998–e101316.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, et al. Microbiota of human breast tissue. Appl Environ Microbiol. 2014;80(10):3007–14.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Wang Z, Wang Q, Zhao J, Gong L, Zhang Y, Wang X, et al. Altered diversity and composition of the gut microbiome in patients with cervical cancer. AMB Express. 2019;9(1):40.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Garrett WS. Cancer and the microbiota. Science. 2015;348(6230):80–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer. 2013;13(11):800–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rajagopala SV, Vashee S, Oldfield LM, Suzuki Y, Venter JC, Telenti A, et al. The human microbiome and Cancer. Cancer Prev Res (Phila). 2017;10(4):226–34.

    Article  PubMed  Google Scholar 

  22. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.

    Article  CAS  PubMed  Google Scholar 

  23. Norenhag J, Du J, Olovsson M, Verstraelen H, Engstrand L, Brusselaers N. The vaginal microbiota, human papillomavirus and cervical dysplasia: a systematic review and network meta-analysis. BJOG. 2020;127(2):171–80.

    Article  CAS  PubMed  Google Scholar 

  24. Wang H, Ma Y, Li R, Chen X, Wan L, Zhao W. Associations of Cervicovaginal Lactobacilli with High-Risk Human Papillomavirus infection, cervical intraepithelial neoplasia, and Cancer: a systematic review and Meta-analysis. J Infect Dis. 2019;220(8):1243–54.

    Article  PubMed  Google Scholar 

  25. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol. 2010;25(9):603–5.

    Article  PubMed  Google Scholar 

  27. Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, Viswanathan M, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355:i4919.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Penson DFKS, Jules A. Evaluation and Treatment of Cryptorchidism. Comparative Effectiveness Review No. 88. Prepared by the Vanderbilt Evidence-based Practice Center under Contract. 2012.

  29. Vali M, Maleki Z, Nikbakht HA, Hassanipour S, Kouhi A, Nazemi S, et al. Survival rate of cervical cancer in Asian countries: a systematic review and meta-analysis. BMC Womens Health. 2023;23(1):671.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Rohatgi A. WebPlotDigitizer. 2020, https://automeris.io/WebPlotDigitizer.

  31. Aydin O, Yassikaya MY. Validity and reliability analysis of the PlotDigitizer Software Program for data extraction from single-case graphs. Perspect Behav Sci. 2022;45(1):239–57.

    Article  PubMed  Google Scholar 

  32. Drevon D, Fursa SR, Malcolm AL. Intercoder Reliability and Validity of WebPlotDigitizer in extracting Graphed Data. Behav Modif. 2017;41(2):323–39.

    Article  PubMed  Google Scholar 

  33. Shi J, Luo D, Weng H, Zeng XT, Lin L, Chu H, et al. Optimally estimating the sample standard deviation from the five-number summary. Res Synth Methods. 2020;11(5):641–54.

    Article  PubMed  Google Scholar 

  34. Liu H, Liang H, Li D, Wang M, Li Y. Association of Cervical Dysbacteriosis, HPV Oncogene expression, and cervical lesion progression. Microbiol Spectr. 2022;10(5):e0015122.

    Article  PubMed  Google Scholar 

  35. Li C, Zhang Z, Yang Y, Liao H. Changes in the cervicovaginal microbiota composition of HPV16-infected patients after clinical treatment. Cancer Med. 2022;11(24):5037–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen Y, Qiu X, Wang W, Li D, Wu A, Hong Z, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jiang L, Li B, Zhang Y, Ma S, Liu C, Liang F, et al. Influence of pelvic intensity-modulated Radiation Therapy with Concurrent Cisplatin-based chemotherapy of Cervical Cancer on the vaginal microbiome. Front Oncol. 2021;11:615439.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kang G-U, Jung D-R, Lee YH, Jeon SY, Han HS, Chong GO, et al. Dynamics of Fecal Microbiota with and without Invasive Cervical Cancer and its application in early diagnosis. Cancers. 2020;12(12):3800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li X, Wu J, Wu Y, Duan Z, Luo M, Li L, et al. Imbalance of vaginal microbiota and immunity: two main accomplices of Cervical Cancer in Chinese women. Int J Women’s Health. 2023;15:987–1002.

    Article  CAS  Google Scholar 

  40. Zeber-Lubecka N, Kulecka M, Lindner B, Krynicki R, Paziewska A, Nowakowski A, et al. Increased diversity of a cervical microbiome associates with cervical cancer. Front Oncol. 2022;12:1005537.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Zhai Q, Zhang W, Zhang Z, Fu Y, Li Y, Wang X, 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.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Zeng W, Liang Y, Zhou J, Lin H, Huang L, He D, et al. Vaginal microecological changes of different degrees of cervical lesions in Hakka women in Meizhou City. J Obstet Gynaecol. 2023;43(1):2186780.

    Article  PubMed  Google Scholar 

  43. Ivanov MK, Brenner EV, Hodkevich AA, Dzyubenko VV, Krasilnikov SE, Mansurova AS, et al. Cervicovaginal-Microbiome analysis by 16S sequencing and real-time PCR in patients from Novosibirsk (Russia) with cervical lesions and several years after Cancer Treatment. Diagnostics. 2023;13(1):140.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ma Y, Li Y, Liu Y, Cao L, Han X, Gao S, et al. Vaginal Microbiome Dysbiosis is Associated with the different cervical Disease Status. J Microbiol. 2023;61(4):423–32.

    Article  CAS  PubMed  Google Scholar 

  45. Stoian IL, Botezatu A, Fudulu A, Ilea CG, Socolov DG. Exploring microbiota diversity in cervical lesion progression and HPV infection through 16S rRNA gene metagenomic sequencing. J Clin Med. 2023;12(15):4979.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zheng X, Hu N, Liu J, Zhao K, Li H, Wang J, et al. Cervicovaginal microbiota disorder combined with the change of cytosine phosphate guanine motif- toll like receptor 9 axis was associated with cervical cancerization. J Cancer Res Clin Oncol. 2023;149(19):17371–81.

    CAS  PubMed  Google Scholar 

  47. Teka B, Yoshida-Court K, Firdawoke E, Chanyalew Z, Gizaw M, Addissie A, et al. Cervicovaginal Microbiota profiles in Precancerous lesions and Cervical Cancer among Ethiopian Women. Microorganisms. 2023;11(4):833.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kang G-U, Jung D-R, Lee YH, Jeon SY, Han HS, Chong GO, et al. Potential Association between vaginal microbiota and cervical carcinogenesis in Korean women: a Cohort Study. Microorganisms. 2021;9(2):294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kwon M, Seo SS, Kim MK, Lee DO, Lim MC. Compositional and functional differences between microbiota and cervical carcinogenesis as identified by shotgun metagenomic sequencing. Cancers. 2019;11(3):309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Sims TT, Biegert GWG, Ramogola-Masire D, Ngoni K, Solley T, Ning MS, et al. Tumor microbial diversity and compositional differences among women in Botswana with high-grade cervical dysplasia and cervical cancer. Int J Gynecol Cancer. 2020;30(8):1151–6.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Sims TT, Colbert LE, Zheng J, Delgado Medrano AY, Hoffman KL, Ramondetta L, et al. Gut microbial diversity and genus-level differences identified in cervical cancer patients versus healthy controls. Gynecol Oncol. 2019;155(2):237–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. So KA, Yang EJ, Kim NR, Hong SR, Lee J-H, Hwang C-S, et al. Changes of vaginal microbiota during cervical carcinogenesis in women with human papillomavirus infection. PLoS ONE. 2020;15(9):e0238705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang Z, Xiao R, Huang J, Qin X, Hu D, Guo E, et al. The diversity of Vaginal Microbiota predicts Neoadjuvant Chemotherapy responsiveness in locally Advanced Cervical Cancer. Microb Ecol. 2022;84(1):302–13.

    Article  CAS  PubMed  Google Scholar 

  54. Wu S, Ding X, Kong Y, Acharya S, Wu H, Huang C, et al. The feature of cervical microbiota associated with the progression of cervical cancer among reproductive females. Gynecol Oncol. 2021;163(2):348–57.

    Article  CAS  PubMed  Google Scholar 

  55. Xie Y, Feng Y, Li W, Zhan F, Huang G, Hu H, et al. Revealing the disturbed vaginal micobiota caused by Cervical Cancer using high-throughput sequencing technology. Front Cell Infect Microbiol. 2020;10:538336.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Xu H, Liu L, Xu F, Liu M, Song Y, Chen J, et al. Microbiome-metabolome analysis reveals cervical lesion alterations. Acta Biochim Biophys Sin. 2022;54(10):1552–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhang Z, Li T, Zhang D, Zong X, Bai H, Bi H, et al. Distinction between vaginal and cervical microbiota in high-risk human papilloma virus-infected women in China. BMC Microbiol. 2021;21(1):90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tsakmaklis A, Vehreschild M, Farowski F, Trommer M, Kohler C, Herter J, et al. Changes in the cervical microbiota of cervical cancer patients after primary radio-chemotherapy. Int J Gynecol Cancer. 2020;30(9):1326–30.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Mitra A, Grossman Biegert GW, Delgado AY, Karpinets TV, Solley TN, Mezzari MP, et al. Microbial Diversity and Composition is Associated with patient-reported toxicity during Chemoradiation Therapy for Cervical Cancer. Int J Radiat Oncol Biol Phys. 2020;107(1):163–71.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Wang Z, Wang Q, Wang X, Zhu L, Chen J, Zhang B, et al. Gut microbial dysbiosis is associated with development and progression of radiation enteritis during pelvic radiotherapy. J Cell Mol Med. 2019;23(5):3747–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Colbert LE, El Alam MB, Wang R, Karpinets T, Lo D, Lynn EJ, et al. Tumor-resident Lactobacillus iners confer chemoradiation resistance through lactate-induced metabolic rewiring. Cancer Cell. 2023;41(11):1945–e6211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang H, Jiang Y, Liang Y, Wei L, Zhang W, Li L. Observation of the cervical microbiome in the progression of cervical intraepithelial neoplasia. BMC Cancer. 2022;22(1):362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Manzanares-Leal GL, Coronel-Martínez J, Rodríguez-Morales M, Bustamante-Montes LP, Sandoval-Trujillo H, Ramírez-Durán N. Changes in the diversity of local cervical bacteria in women with cervical cancer receiving antineoplastic treatment. J Res Med Sci. 2021;26(1):56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sobhani I, Bergsten E, Couffin S, Amiot A, Nebbad B, Barau C, et al. Colorectal cancer-associated microbiota contributes to oncogenic epigenetic signatures. Proc Natl Acad Sci U S A. 2019;116(48):24285–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Amabebe E, Anumba DOC. Female gut and genital Tract Microbiota-Induced Crosstalk and Differential effects of short-chain fatty acids on Immune Sequelae. Front Immunol. 2020;11:2184.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Nunn KL, Forney LJ. Unraveling the dynamics of the human vaginal microbiome. Yale J Biol Med. 2016;89(3):331–7.

    PubMed  PubMed Central  Google Scholar 

  67. Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host Microbe. 2011;10(4):324–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, et al. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med. 2012;10:253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas. 2017;103:45–53.

    Article  CAS  PubMed  Google Scholar 

  70. Amini-Salehi E, Samethadka Nayak S, Maddineni G, Mahapatro A, Keivanlou MH, Soltani Moghadam S, et al. Can modulation of gut microbiota affect anthropometric indices in patients with non-alcoholic fatty liver disease? An umbrella meta-analysis of randomized controlled trials. Ann Med Surg (Lond). 2024;86(5):2900–10.

    Article  PubMed  Google Scholar 

  71. Naghipour A, Amini-Salehi E, Orang Gorabzarmakhi M, Shahdkar M, Fouladi B, Alipourfard I, et al. Effects of gut microbial therapy on lipid profile in individuals with non-alcoholic fatty liver disease: an umbrella meta-analysis study. Syst Rev. 2023;12(1):144.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Amini-Salehi E, Hassanipour S, Keivanlou MH, Shahdkar M, Orang Goorabzarmakhi M, Vakilpour A, et al. The impact of gut microbiome-targeted therapy on liver enzymes in patients with nonalcoholic fatty liver disease: an umbrella meta-analysis. Nutr Rev. 2024;82(6):815–30.

    Article  PubMed  Google Scholar 

  73. Wang J, Zhu N, Su X, Gao Y, Yang R. Gut-microbiota-derived metabolites maintain gut and systemic Immune Homeostasis. Cells. 2023;12(5).

  74. Zhao LY, Mei JX, Yu G, Lei L, Zhang WH, Liu K, et al. Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduct Target Ther. 2023;8(1):201.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Algrafi AS, Jamal AA, Ismaeel DM. Microbiota as a New Target in Cancer Pathogenesis and Treatment. Cureus. 2023;15(10):e47072.

    PubMed  PubMed Central  Google Scholar 

  76. Gerassy-Vainberg S, Blatt A, Danin-Poleg Y, Gershovich K, Sabo E, Nevelsky A, et al. Radiation induces proinflammatory dysbiosis: transmission of inflammatory susceptibility by host cytokine induction. Gut. 2018;67(1):97–107.

    Article  CAS  PubMed  Google Scholar 

  77. Panebianco C, Adamberg K, Jaagura M, Copetti M, Fontana A, Adamberg S, et al. Influence of gemcitabine chemotherapy on the microbiota of pancreatic cancer xenografted mice. Cancer Chemother Pharmacol. 2018;81(4):773–82.

    Article  CAS  PubMed  Google Scholar 

  78. Acero Brand FZ, Suter N, Adam JP, Faulques B, Maietta A, Soulières D, et al. Severe immune mucositis and esophagitis in metastatic squamous carcinoma of the larynx associated with pembrolizumab. J Immunother Cancer. 2018;6(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Chase D, Goulder A, Zenhausern F, Monk B, Herbst-Kralovetz M. The vaginal and gastrointestinal microbiomes in gynecologic cancers: a review of applications in etiology, symptoms and treatment. Gynecol Oncol. 2015;138(1):190–200.

    Article  PubMed  Google Scholar 

  80. Montassier E, Gastinne T, Vangay P, Al-Ghalith GA, Bruley des Varannes S, Massart S, et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther. 2015;42(5):515–28.

    Article  CAS  PubMed  Google Scholar 

  81. Lin XB, Farhangfar A, Valcheva R, Sawyer MB, Dieleman L, Schieber A, et al. The role of intestinal microbiota in development of irinotecan toxicity and in toxicity reduction through dietary fibres in rats. PLoS ONE. 2014;9(1):e83644.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science. 2010;330(6005):831–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zhou CB, Zhou YL, Fang JY. Gut microbiota in Cancer Immune Response and Immunotherapy. Trends Cancer. 2021;7(7):647–60.

    Article  CAS  PubMed  Google Scholar 

  84. Maduro JH, Pras E, Willemse PH, de Vries EG. Acute and long-term toxicity following radiotherapy alone or in combination with chemotherapy for locally advanced cervical cancer. Cancer Treat Rev. 2003;29(6):471–88.

    Article  CAS  PubMed  Google Scholar 

  85. Fessler J, Matson V, Gajewski TF. Exploring the emerging role of the microbiome in cancer immunotherapy. J Immunother Cancer. 2019;7(1):108.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Martin DH, Marrazzo JM. The vaginal microbiome: current understanding and future directions. J Infect Dis. 2016;214(Suppl 1):S36–41.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Hickey RJ, Zhou X, Pierson JD, Ravel J, Forney LJ. Understanding vaginal microbiome complexity from an ecological perspective. Transl Res. 2012;160(4):267–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Mitra A, MacIntyre DA, Ntritsos G, Smith A, Tsilidis KK, Marchesi JR, et al. The vaginal microbiota associates with the regression of untreated cervical intraepithelial neoplasia 2 lesions. Nat Commun. 2020;11(1):1999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Zevin AS, Xie IY, Birse K, Arnold K, Romas L, Westmacott G, et al. Microbiome composition and function drives Wound-Healing impairment in the female genital tract. PLoS Pathog. 2016;12(9):e1005889.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Fernandes JV, Cobucci RN, Jatobá CA, Fernandes TA, de Azevedo JW, de Araújo JM. The role of the mediators of inflammation in cancer development. Pathol Oncol Res. 2015;21(3):527–34.

    Article  CAS  PubMed  Google Scholar 

  91. De Marco F. Oxidative stress and HPV carcinogenesis. Viruses. 2013;5(2):708–31.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Doorbar J. Molecular biology of human papillomavirus infection and cervical cancer. Clin Sci (Lond). 2006;110(5):525–41.

    Article  CAS  PubMed  Google Scholar 

  93. Muñoz N, Bosch FX, de Sanjosé S, Herrero R, Castellsagué X, Shah KV, et al. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med. 2003;348(6):518–27.

    Article  PubMed  Google Scholar 

  94. Steenbergen RD, Snijders PJ, Heideman DA, Meijer CJ. Clinical implications of (epi)genetic changes in HPV-induced cervical precancerous lesions. Nat Rev Cancer. 2014;14(6):395–405.

    Article  CAS  PubMed  Google Scholar 

  95. Zhang W, Yin Y, Jiang Y, Yang Y, Wang W, Wang X, et al. Relationship between vaginal and oral microbiome in patients of human papillomavirus (HPV) infection and cervical cancer. J Transl Med. 2024;22(1):396.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Onderdonk AB, Delaney ML, Fichorova RN. The human microbiome during bacterial vaginosis. Clin Microbiol Rev. 2016;29(2):223–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Fethers K, Twin J, Fairley CK, Fowkes FJ, Garland SM, Fehler G, et al. Bacterial vaginosis (BV) candidate bacteria: associations with BV and behavioural practices in sexually-experienced and inexperienced women. PLoS ONE. 2012;7(2):e30633.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Mitra A, MacIntyre DA, Paraskevaidi M, Moscicki AB, Mahajan V, Smith A, et al. The vaginal microbiota and innate immunity after local excisional treatment for cervical intraepithelial neoplasia. Genome Med. 2021;13(1):176.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Hemmerling A, Harrison W, Schroeder A, Park J, Korn A, Shiboski S, et al. Phase 2a study assessing colonization efficiency, safety, and acceptability of Lactobacillus crispatus CTV-05 in women with bacterial vaginosis. Sex Transm Dis. 2010;37(12):745–50.

    Article  PubMed  Google Scholar 

  100. Ngugi BM, Hemmerling A, Bukusi EA, Kikuvi G, Gikunju J, Shiboski S, et al. Effects of bacterial vaginosis-associated bacteria and sexual intercourse on vaginal colonization with the probiotic Lactobacillus crispatus CTV-05. Sex Transm Dis. 2011;38(11):1020–7.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65.

    Article  CAS  PubMed  Google Scholar 

  102. Wilkinson EM, Ilhan ZE, Herbst-Kralovetz MM. Microbiota-drug interactions: impact on metabolism and efficacy of therapeutics. Maturitas. 2018;112:53–63.

    Article  CAS  PubMed  Google Scholar 

  103. Kurita A, Kado S, Matsumoto T, Asakawa N, Kaneda N, Kato I, et al. Streptomycin alleviates irinotecan-induced delayed-onset diarrhea in rats by a mechanism other than inhibition of β-glucuronidase activity in intestinal lumen. Cancer Chemother Pharmacol. 2011;67(1):201–13.

    Article  CAS  PubMed  Google Scholar 

  104. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.

    Article  CAS  PubMed  Google Scholar 

  106. Colbert LE, et al. The gut and cervical microbiome promote immune activation and response to chemoradiation in cervical cancer. Cancer Cell. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.2139/ssrn.3199993.

    Article  Google Scholar 

  107. Clarridge JE 3. Impact of 16S rRNA gene sequence analysis for identification of bacteria on clinical microbiology and infectious diseases. Clin Microbiol Rev. 2004;17(4):840–62. table of contents.

  108. Woo PC, Lau SK, Teng JL, Tse H, Yuen KY. Then and now: use of 16S rDNA gene sequencing for bacterial identification and discovery of novel bacteria in clinical microbiology laboratories. Clin Microbiol Infect. 2008;14(10):908–34.

    Article  CAS  PubMed  Google Scholar 

  109. Wensel CR, Pluznick JL, Salzberg SL, Sears CL. Next-generation sequencing: insights to advance clinical investigations of the microbiome. J Clin Invest. 2022;132(7).

  110. Poulsen CS, Ekstrøm CT, Aarestrup FM, Pamp SJ. Library Preparation and sequencing platform introduce Bias in Metagenomic-based characterizations of Microbiomes. Microbiol Spectr. 2022;10(2):e0009022.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Health Commission of Sichuan Province Medical Science and Technology Program (24QNMP038), the Chengdu Technology Bureau (2024-YF05-02230-SN), and the Health Commission of Sichuan Province (2024–803).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, J.L. and Q.W.; methodology, Q.W. and M.C.; screening and assessment of eligible studies, M.C. and S.W.; data extraction, S.W., Y.M., X.L., and J.F.; quality assessment, Y.M. X.L., and J.F.; original draft preparation, Q.W.; supervision and funding acquisition, J.L. and M.C. All the authors reviewed and agreed to the final version of the manuscript.

Corresponding authors

Correspondence to Jinyi Lang or Meihua Chen.

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.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wen, Q., Wang, S., Min, Y. et al. Associations of the gut, cervical, and vaginal microbiota with cervical cancer: a systematic review and meta-analysis. BMC Women's Health 25, 65 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12905-025-03599-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12905-025-03599-1

Keywords