- Research
- Open access
- Published:
Comprehensive analysis of the interaction microbiome and prostate cancer: an initial exploration from multi-cohort metagenome and GWAS studies
Journal of Translational Medicine volume 23, Article number: 130 (2025)
Abstract
Introduction
Prostate cancer is one of the most common cancers in the United States with a high mortality rate. In recent years, the traditional opinion about prostate microbiome was challenged. Although there still are some arguments, an escalating number of researchers are shifting their focus toward the microbiome within the prostate tumor environment.
Methods
We mined the data of the microbiome extracted from the metagenome, and it offers a broader taxonomic coverage and accurate functional profiling. We used Kraken2, a mapping tool, to mine the gut microbiota of prostate cancer patients. A two-sample Mendelian Randomization was conducted to reflect the association between gut microbiome and cancer.
Results
In the study, we found the consistency of the special intratumor microbiome of both non-metastatic tumors and metastatic tumors. And we dig the gut microbiome in patients with different treatments. We found that some microbiotas may be associated with prostate cancer progression and a special microbiome in metastatic prostate cancer may exist. The anti-androgen therapy can significantly change both the intratumor and gut microbiome.
Conclusion
With the progression and metastasis of prostate cancer, some intratumor microbiome changes. And anti-androgen influences both the intratumor and gut microbiome. Our discovery may help researchers further understand the progression, metastasis, and resistance of prostate cancer from the perspective of microbiome level.
Introduction
In 2020, there were over 1.4 million newly diagnosed prostate cancer patients, and more than 375 thousand patients died of this terrible disease [1]. In America, the prostate is the most common cancer in men, and the mortality of prostate cancer is just lower than lung cancer [2]. In Asia, this situation is also not optimistic. In recent years, the prostate cancer incidence has still been increasing [3]. Exploration of the mechanism of prostate cancer formation and progression is becoming more and more important.
In the past, the urine was thought sterile. This stereotypical impression made the urologist surgeons and researchers pay no attention to the microbiome in prostate cancer. However, the association between microbiome and prostate cancer is attracting researchers’ attention, especially after a study demonstrating the existence of an intratumoral microbiome was published in 2020 in Science [4]. This means researchers are paying more attention to the microbiome beyond gut diseases. Recent research has indicated that there is an association between microbiome and the incidence and progression of prostate cancer. The intestinal microbiota of patients could participate in metabolism and lead to resistance [5]. And some studies reported the role of microbiota in signaling [6, 7]. The signature of intratumoral microbiota was also reported, though the results are not consistent [8,9,10,11]. To further explore what role the microbiome plays in the progression, metastasis, and resistance in prostate cancer, it is important to broaden the source of the microbiome information. Due to the complexity of microbiota, bioinformatics plays an important role in identifying the characteristics and relevance of the microbiome in patients. As for metagenomics, which analyzes microbiota from the environment directly, it offers a broader taxonomic coverage and accurate functional profiling [12]. The data of microbiome extracted from metagenome were recently used to analysis with 16 S rRNA sequencing data in some research and performed well [13, 14]. We gained the signature of the microbiome in prostate cancer patients from the published articles and used similar tools to analyze the gut microbiome [15, 16]. Kraken2, a bioinformatics tool employed for the mapping and analysis of microbiota within the context of prostate cancer patients, is fast and precise while minimizing the consumption of computing resources [16]. This software has been extensively leveraged across a spectrum of prior research endeavors, thereby accruing a robust body of validation and scholarly recognition. Its user-friendly configuration and the facile interpretability of its output render it a highly accessible asset for subsequent investigative pursuits [17]. Kraken2 was also used in the intratumor microbiome data we have extracted in recent studies [18], providing us with valuable insights into this complex biological landscape.
With the development of bioinformatics, more tools can be used to study the risk factors of prostate cancer at a higher dimension. Mendelian randomization (MR) is a new tool evaluating the causal effects using genetic variants. Nonmodifiable genetic instruments ensure lifelong exposure, which can avoid both research bias and confounding environmental factors [19]. In this study, a two-sample MR is also used to evaluate the association between gut microbiome and prostate cancer. The results may offer a new vision of the influence of microbiota in the formation and progression of prostate cancer.
Methods
Intratumoral microbiome data
We searched Pubmed and extracted microbiome data from reliable studies. A study mining microbiome of 4164 metastases from the Hartwig Medical Foundation was included [18]. The Hartwig Medical Foundation is a project under the Center for Personalized Cancer Treatment (CPCT). Its goal was to systemically collect clinical data of patients with metastatic cancer while collecting biopsy metastatic lesions for whole genome sequencing. The data of prostate cancer patients was downloaded and screened using R packages “MicrobeDS” and “phyloseq”.
TCGA, a landmark cancer genomics program, molecularly characterized over 20,000 primary cancers and matched normal samples. With comprehensive clinical data, it is widely used in many studies about cancer. A study extracting microbiome from The Cancer Genome Atlas Program (TCGA) using miRNA data was also included in [20]. The detailed microbiome data can be downloaded from the web link in their article (http://bic.jhlab.tw/).
Gut microbiome data
We searched the gut metagenome data from the National Center of Biotechnology Information (NCBI) BioProject database (https://www.ncbi.nlm.nih.gov/bioproject). The project names are PRJDB10718, PRJDB9379, PRJNA1077793. The MiSeq (Illumina) system was used to obtain 16s RNA. The detailed information was shown in the Table 1. We obtained the raw sequence data from Sequence Read Archive (SRA) database (https://www.ncbi.nlm.nih.gov/sra).
Microbial profiling pipeline
Preprocession
The workflow of bioinformatics was performed using default parameters unless we stated otherwise. All the whole metagenome sequencing (WMS) or 16 S rRNA reads were preprocessed using package “fastp”, which filter out low quality, short reads, and trim reads in front and tail [21]. Program “fastQC” was used to spot potential problems in sequencing datasets.
Aligning sequencing reads
Bowtie 2, a fast and memory-efficient tool, was used for aligning sequencing reads to human reference sequences [22, 23]. We used genome assembly GRCh38 as human reference sequences and downloaded pre-built genome indexes “Human / GRCh38 no-alt analysis set” from the official website (https://bowtie-bio.sourceforge.net/bowtie2/index.shtml). And we collected all the sequencing that did not align to the human genome (GRCh38). “Samtools” was used to filter reads that did not map successfully and produce a binary alignment/map (BAM) file for the following analysis [24].
Reads analysis
16 S rRNA reads and metagenome reads were analyzed with Kraken2, a rapid and multithreading axonomic sequence classifier that assigns taxonomic labels to sequences [16]. It can get on their website (https://benlangmead.github.io/aws-indexes/k2). A Kraken2 reference database named “PlusPF” was access on 30 June 2024. It is consistent of archaea, bacteria, viral, plasmid, human, UniVecCore, protozoa and fungi. Microbiota reads were extracted using Kraken2 from WMS reads or 16s RNA reads. After Kraken2 profiling, Bracken2 software was used to re-estimate genus-level abundances, applying a Bayesian model [15].
Depletion of known contaminants
To correct for false detection of microbiota, we adopted published list of possibility genera and removed both likely contaminants and mixed evidence contaminants [25]. And we searched studies about microbiome in prostate cancer and adopted reported microbiota into further consideration.
Bioinformatics: microbial analysis
Alpha and beta diversity estimations
Alpha diversity reflected the richness and diversity of the microbiome within the sample. The R package “vegan” (version: 2.6.6.1) was used to calculate the alpha diversity estimations. The Shannon and Simpson index were calculated to evaluate the richness and evenness of the microbiome [26, 27]. The Chao index, ACE index, and Pielou evenness were also calculated. The Chao index and ACE index estimate the richness of the sample or community [28, 29]. Pielou’s evenness, the ratio of the actual Shannon index of a community to the maximum Shannon index, measures diversity along with species richness and is the most common measure of evenness [30]. An online tool was used for the figure graphing (https://www.genescloud.cn/chart/ChartOverview).
And packages “ggplot2” (version: 3.5.1), “ggprism” (version: 1.0.5) and “vegan” (version: 2.6.6.1) were used for beta diversity estimations and graph. The Bray-Curtis distance [31] and PCoA [32], reflecting the microbiome profiles, were shown in figure.
Microbial analysis
The public microbiome data of prostate cancer can get using R package “MicrobeDS”. To analysis the consistent of microbiome, the packages “pheatmap” (version: 1.0.12) and “phyloseq” (version: 1.46.0) were used. Heatmap and barplot were used to reflect the component. A univariate cox regression analyses was conducted, and we tried to identify special cancer species using “survival” (version 3.7.0) and “survminer” (version: 0.4.9).
Survival analysis
We got the clinical data for survival analysis from TCGA database (https://portal.gdc.cancer.gov) to identify microbe signatures associated with prognosis. Kaplan-Meier (KM) curves were plotted by the “survminer” (version: 0.4.9) and it reflected the influence of these special microbiota in prognostic of prostate cancer. The Maximally Selected Rank Statistics was used for KM curves plotting.
MR
The data resources were obtained from MRC IEU OpenGWAS (https://gwas.mrcieu.ac.uk/; version: v8.5.1–2024-07-17), developed at the MRC Integrative Epidemiology Unit at the University of Bristol. The GWAS study we included was ieu-b-85 (Prostate cancer). The FinnGen (https://www.finngen.fi/fi/) project, commenced in autumn 2017, was also included. It was a public-private research collected from the Finnish Biobank and the Finnish Health Registry and included around 500,000 participants [33]. The majority of the FinnGen data in this study originated from Round 10. The data of gut microbiota can be accessed from GCST90032172 to GCST90032644, and the detail has been reported in a published article [34]. All of the p values are two tails. The R software (version 4.3.1) with the Two Sample MR package was used in the analysis. Figures were plotted by https://www.bioinformatics.com.cn (last accessed on 20 June 2024), an online platform for data analysis and visualization.
Result
Analysis of intratumoral microbiome
The microbiome of prostate cancer
In the microbiome data of TCGA, we explored the difference between the tumor tissue and non-tumor tissue (non-malignant adjacent prostate samples) [35]. Just as Fig. 1 showed, within the microbiome of TCGA-PRAD, the Proteobacteria emerged as the most abundant phylum. High abundance levels were also observed for Firmicutes, Actinobacteria, and Bacteroidetes. Among the five most abundant genera identified in the TCGA-PRAD microbiome were Bacillus, Pseudomonas, Paenibacillus, Acinetobacter, and Corynebacterium. Subsequently, we estimated the microbiome within prostate tumors using a variety of methodologies. We observed a significant difference in alpha-diversity (p < 0.05). The Shannon index, Simpson index, and Pielou’s evenness all exhibited lower in the tumor group, which meant community richness and community evenness were lower (Fig. 2A). In the beta-diversity analysis results, we discovered significant differences in intratumoral microbiome profiles (p < 0.05) (Fig. 2B and C).
(A) The alpha diversity of microbiome in TCGA-PRAD, including Shannon index, Simpson index, Richness, Chao, Obs, and Pielou’s evenness. (B) The PCoA of TCGA-PRAD microbiome group by tumor tissue and non-tumor tissue. 3 (C) The Bray-Curtis distance of TCGA-PRAD microbiome group by tumor tissue and non-tumor tissue
An analysis utilizing linear discriminant analysis effect size (LEfSe) revealed distinct microbiotas at the phylum, family, and genus level (p < 0.05) (Fig. 3A, B and C). Searching the List of Prokaryotic names with Standing in Nomenclature (LPSN) [36] and deleting low abundant microbiome, we included some possible families (Comamonadaceae, Cystobacteraceae, Enterobacteriaceae, Mycobacteriaceae, Paenibacillaceae and Peptostreptococcaceae) and genus (Mycobacterium, Paenibacillus, Peptoclostridium, Saccharomonospora, Salmonella and Alicyclobacillus) in following analysis. The Kaplan-Meier curves revealed a significant impact of the intratumor microbiome on the progression of prostate cancer, with differences observed at both the genus and phylum levels, as illustrated in Fig. 3D and E. Subgroup analysis of the PSA value ( < = 0.2 ng/ml and > 0.2 ng/ml) and tumor pathology stage (T1/T2 and T3/T4) were shown in Supplemental Fig. 1. We also conducted a univariate Cox regression analysis on the highly abundant microbiota and identified that Paenibacillus, Mycobacterium and Streptococcus may be associated with biochemical recurrence. The detailed results of this analysis are presented in Supplemental Table 1.
(A) The LDA EFfect Size (LEfSe) analysis of microbiota group by tumor tissue and non-tumor tissue at phylum level (p < 0.05). (B) The LEfSe analysis of microbiota group by tumor tissue and non-tumor tissue at family level (p < 0.05). (C) The LEfSe analysis of microbiota between tumor and normal tissue at genus level (p < 0.05). (D) The positive result of Kaplan-Meier Survival Analysis group by microbiotas at family level. (E) The positive result of Kaplan-Meier Survival Analysis group by microbiotas at genus level
The microbiome of metastatic cancer
In metastatic prostate cancer, we discovered that Staphylococcaceae, Streptococcaceae, Pseudomonadaceae, and Enterobacteriaceae were the most abundant families at the family level (Fig. 4). And it was discovered that Staphylococcus, Streptococcus, Lactobacillus, Pseudomonas, Clostridium, Klebsiella, and Bacteroides were rich in metastatic prostate cancer (Supplemental Fig. 2). We analyzed the intratumoral microbiota of metastatic prostate cancer but found no significant difference in alpha-diversity or beta-diversity among the samples at different biopsy sites (Fig. 5A, B and C).
(A) The alpha diversity of microbiome in metastasis prostate cancer, including Shannon index, Simpson index, Richness, Chao, Obs, and Pielou’s evenness, group by biopsy site. (B) The PCoA of metastasis prostate cancer microbiome group by tumor tissue and non-tumor tissue, group by biopsy site. (C) The Bray-Curtis distance of metastasis prostate cancer microbiome group by biopsy site
Then we also conducted a comprehensive analysis of microbiota variations under various treatment modalities and discovered that anti-androgen therapy (androgen deprivation therapy and hormonal therapy) and immunotherapy can significantly alter the intra-cellular microbiome of patients (Fig. 6A and B, and 6C). To further elucidate the distinct within the metastatic prostate cancer microbiome, the LEfSe study was conducted. Notably, our analysis revealed that androgen deprivation therapy (ADT) and radiation therapy significantly modulate the intratumoral microbiome of metastatic prostate cancer patients. All the results were adjusted by False discovery rate (FDR) and the significant result lists were shown in Fig. 6D and E. The relative abundance of Olsenella, Myroides, Arcobacter, Collinsella and Parascardovia was significantly elevated compared to other groups. Within the cohort receiving radiation therapy, Leptotrichia, Olsenella, Parascardovia, Brachytbacterium, and Collinsella were higher. Moreover, as an exploration study, we upload the result of the unadjusted p-value in Supplemental Fig. 3.
(A) The alpha diversity of microbiome in metastasis prostate cancer, including Shannon index, Simpson index, Richness, Chao, Obs, and Pielou’s evenness, group by treatment type. (B) The PCoA of metastasis prostate cancer microbiome group by tumor tissue and non-tumor tissue, group by treatment type. (C) The Bray-Curtis distance of metastasis prostate cancer microbiome group by treatment type. (D) The LEfSe analysis of microbiota between androgen deprivation patients and other treatments patients. (E) The LEfSe analysis of microbiota between nuclear treatment patients and other treatments patients
MR
The GWAS ID and details of the included studies was shown in Table 1. The MR outcome of ieu-b-85 was depicted in Fig. 7, Supplemental Fig. 4. In the two-sample MR analysis of ieu-b-85, 19 kinds of gut microbiota were identified to be correlated with prostate cancer. 9 of the identified gut microbiota species were associated with an increased risk of prostate cancer, while 10 were correlated with a decreased risk. However, none of these associations remained statistically significant after adjusting by FDR. The MR analysis result of the Finn database was shown in Supplemental Fig. 5.
Analysis of gut microbiome
In the previous analysis, we found distinct microbial profiles in the patients who received some specific therapeutic. Considering the multi-source of the microbiota in the tumor, comparing the microbiota composition outside the tumor may help us identify the key microbiota and provide new insights into prostate cancer. Consequently, we applied kraken2, a sequence classifier, to explore the gut microbiome of prostate cancer patients.
We initially investigated the gut microbiome in suspected prostate cancer patients. A bar plot was used to describe the consistency of the gut microbiome, presented in Fig. 8. Our previous study indicated that anti-androgen therapy and nuclear therapy may influence the microbiome of patients. Subsequently, we explored the gut microbiota in the patients with radiation therapy and anti-androgen therapy. However, no statistically significant differences were observed after applying the FDR correction to the p-values. As an exploration study, we tried to analyze the data based on the unadjusted p-values. And the difference microbiotas were depicted in Fig. 9.
Discussion
The relationship between the microbiome and prostate cancer is still on debate. Till now, few trials have been conducted, and whether a special prostate microbiome exists is still being doubted [37]. Although the urinary was thought sterile in tradition, some special microbiotas were still found in prostate cancer. In the majority of studies preceding this discussion, Propionibacterium was identified as the dominant microorganism [11, 38, 39], which is also reported associated with prostate inflammation. As for normal microbiota, Streptococcaceae was found with a lower abundance in tumor tissue while Staphylococcaceae was higher [11].However, most of the findings were obtained by traditional nucleic acid amplification tests like PCR or 16 S rRNA. The amplification bias may distort the bacterial composition and quantify, and it also could not capture viruses [9]. In recent years, the metagenome has entered people’s field of vision. Some studies using metagenome analysis tools reported some low-abundance microbiota such as Acinetobacterter, Pseudomonas, Firmicutes and Actinobacteria [9, 10]. Tumorigenic including cytomegalovirus and human papillomavirus were also reported [10]. The newly discovered low-abundance microbiota may help us understand more about the microenvironment of cancers and their interaction.
In our study of the intratumor microbiome, we classified the change of alpha and beta diversity between tumor and normal tissue. The community richness and community evenness were lower in tumor tissue, and this phenomenon has been observed in many other types of tumor [40,41,42]. A possible explanation was that injury to the prostatic epithelium or reductions androgen regulated secretory capability promote pathogenic organisms [43]. It can also be an explanation for the observed disparities of beta diversity. The opportunistic bacteria immigrate to the tumor microenvironment (TME) and inhibit abundant indigenous microbiota.
Although plenty of studies identified the microbiome in primary prostate cancer studies, the data of metastatic prostate cancer was still absent. We extracted the microbiome data from a recent pan-cancer study and found that Staphylococcus and Streptococcus were the highest genus, but they existed on normal human skin and were normal environmental pollutants. We also identified some genera, such as Lactobacillus, Pseudomonas, and Clostridium, which may associated with metastasis of cancer, which has been reported in other cancers. Some past studies may be helpful to explain the specific mechanism. Clostridium, plays an important role in the cancer progression through androgen receptor signaling [44]. Bacteroides, were found associated with a greater risk of prostate cancer in a study of the gut microbiome [45]. Streptococcus and Lactobacillus, were also found enriched in the fecal samples of CRPC patients [5]. Interestingly, we found no difference among the samples from different biopsy sites. And we supposed that a special prostate microbiome may exist and keep its special features even if it spreads to different sites. Then we tried to explore the aftermath of distinct treatment types. Anti-androgen therapy, a first-line treatment of advanced prostate cancer, showed a great benefit to patients. But unfortunately, almost all the patients receive the hormonal therapy or androgen deprivation therapy will advance to castration-resistant prostate cancer (CRPC) within 3 years [46]. How to maintain the sensitivity of prostate cancer to anti-androgen therapy is a great challenge for us. In this study, we found Olsenella, Arcobacter, Parascardovia, and other microbiotas have higher abundance after hormonal treatment. In fact, it has been found endocrine resistance by gut bacteria androgen biosynthesis [5]. In intratumor microbiota, some bacteria which can synthesize androgen may also increase as an alternative. Also, the lack of androgen can change the immunity statement, and higher androgen levels could suppress antitumor immunity [47]. Also, the intratumor microbiome could reflect the metabolism, the change of abundance shows the change of environment in cancer just as a nonliving material. And the little change may not be detected using normal instruments.
Nowadays, the interaction of whole body microbiome is getting researchers attention [48]. As for the intratumor microbiome, four potential sources have been proposed, including hematogenous spread, lymphatic drainage, normal adjacent tissue (NAT), and mucosal barriers [49]. We also study the gut microbiome and try to demonstrate the links between the gut and tumor microbiota. Both the analysis of 16sRNA microbiome data and the MR study didn’t show significance after the p value adjusting by FDR. As an exploratory study, we decided to analyze the original p value.
Although some studies suggest gut microbiota plays a role in prostate cancer, but not much study has studied the mechanism under the association. Recent studies offer a possible vision. A major metabolite of microbiota called short chain fatty acids can activate insulin-like growth factor 1 (IGF-1) production and IGF-1 signaling pathway promotes the growth of prostate cancer [7]. The interleukin-6 (IL6) signaling pathway and activator of transcription 3 (STAT3) axis activated by LPS may also promote cancer growth. A former study has found that in CRPC patients, some gut microbiota may produce a bacterial enzyme that synthesizes androgenic steroids and lead to endocrine treatment resistance [5]. And other studies reported phylum Firmicutes, like Lactobacillus, can be responsible for testosterone regulation in patients [50, 51].
Conclusion
In summary, we offered a new vision of the microbiome in prostate cancer. We discovered the possibility of special prostate microbiota existence. The anti-androgen treatment can also influence both the intratumor and gut microbiome. This discovery could help researchers further understand the mechanism of progression, metastasis and treatment.
Data availability
All the human data we used in this article is open to access. And the detail is declared in the article.
Abbreviations
- RNA:
-
Ribonucleic acid
- MR:
-
Mendelian Randomization
- CPCT:
-
Center for Personalized Cancer Treatment
- TCGA:
-
The Cancer Genome Atlas Program
- NCBI:
-
National Center of Biotechnology Information
- SRA:
-
Sequence Read Archive
- WMS:
-
Whole metagenome sequencing
- BAM:
-
Binary Alignment/Map
- KM:
-
Kaplan-Meier
- LEfSe:
-
Linear discriminant analysis effect size
- LPSN:
-
The List of Prokaryotic names with Standing in Nomenclature
- ADT:
-
Androgen deprivation therapy
- FDR:
-
False discovery rate
- GWAS:
-
Genome-Wide Association Studies
- PCR:
-
Polymerase Chain Reaction
- NAT:
-
Normal adjacent tissue
- IGF-1:
-
Insulin-like growth factor 1
- IL6:
-
Interleukin-6
- LPS:
-
Lipopolysaccharides
- CRPC:
-
Castration-Resistant Prostate Cancer
- PCoA:
-
Principal Co-ordinates Analysis
References
Gandaglia G, Leni R, Bray F, Fleshner N, Freedland SJ, Kibel A, et al. Epidemiology and Prevention of prostate Cancer. Eur Urol Oncol. 2021;4:877–92.
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7–30.
Zhu Y, Mo M, Wei Y, Wu J, Pan J, Freedland SJ, et al. Epidemiology and genomics of prostate cancer in Asian men. Nat Rev Urol. 2021;18:282–301.
Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368:973–80.
Pernigoni N, Zagato E, Calcinotto A, Troiani M, Mestre RP, Calì B, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374:216–24.
Liu Y, Yang C, Zhang Z, Jiang H. Gut microbiota dysbiosis accelerates prostate Cancer progression through increased LPCAT1 expression and enhanced DNA repair pathways. Front Oncol. 2021;11:679712.
Matsushita M, Fujita K, Hayashi T, Kayama H, Motooka D, Hase H, et al. Gut microbiota–derived short-chain fatty acids promote prostate Cancer Growth via IGF1 Signaling. Cancer Res. 2021;81:4014–26.
Yow MA, Tabrizi SN, Severi G, Bolton DM, Pedersen J et al. Australian Prostate Cancer BioResource,. Characterisation of microbial communities within aggressive prostate cancer tissues. Infect Agent Cancer. 2017;12:4.
Feng Y, Ramnarine VR, Bell R, Volik S, Davicioni E, Hayes VM, et al. Metagenomic and metatranscriptomic analysis of human prostate microbiota from patients with prostate cancer. BMC Genomics. 2019;20:146.
Banerjee S, Alwine JC, Wei Z, Tian T, Shih N, Sperling C, et al. Microbiome signatures in prostate cancer. Carcinogenesis. 2019;40:749–64.
Cavarretta I, Ferrarese R, Cazzaniga W, Saita D, Lucianò R, Ceresola ER, et al. The Microbiome of the prostate Tumor Microenvironment. Eur Urol. 2017;72:625–31.
Bharti R, Grimm DG. Current challenges and best-practice protocols for microbiome analysis. Brief Bioinform. 2019;22:178–93.
Herzog EL, Kreuzer M, Zinkernagel MS, Zysset-Burri DC. Challenges and insights in the exploration of the low abundance human ocular surface microbiome. Front Cell Infect Microbiol. 2023;13:1232147.
Mannion A, Sheh A, Shen Z, Dzink-Fox J, Piazuelo MB, Wilson KT et al. Shotgun metagenomics of gastric biopsies reveals compositional and functional microbiome shifts in high- and low-gastric-Cancer-risk populations from Colombia, South America. Gut Microbes. 2023;15:2186677.
Lu J, Breitwieser FP, Thielen P, Salzberg SL. Bracken: estimating species abundance in metagenomics data. PeerJ Comput Sci. 2017;3:e104.
Wood DE, Lu J, Langmead B. Improved metagenomic analysis with Kraken 2. Genome Biol. 2019;20:257.
Shahzad M, Saeedullah A, Khan MS, Ahmad HA, Iddrissu I, Andrews SC. 16S rRNA gene amplicon sequencing data from the gut microbiota of adolescent Afghan refugees. Data Brief. 2024;55:110636.
Battaglia TW, Mimpen IL, Traets JJH, van Hoeck A, Zeverijn LJ, Geurts BS, et al. A pan-cancer analysis of the microbiome in metastatic cancer. Cell. 2024;187:2324–e233519.
Sekula P, Del Greco MF, Pattaro C, Köttgen A. Mendelian randomization as an Approach to assess causality using Observational Data. J Am Soc Nephrol JASN. 2016;27:3253–65.
Chen K-P, Hsu C-L, Oyang Y-J, Huang H-C, Juan H-F. BIC: a database for the transcriptional landscape of bacteria in cancer. Nucleic Acids Res. 2023;51:D1205–11.
Chen S. Ultrafast one-pass FASTQ data preprocessing, quality control, and deduplication using fastp. iMeta. 2023;2:e107.
Langmead B, Wilks C, Antonescu V, Charles R. Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics. 2019;35:421–32.
Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–9.
Danecek P, Bonfield JK, Liddle J, Marshall J, Ohan V, Pollard MO, et al. Twelve years of SAMtools and BCFtools. GigaScience. 2021;10:giab008.
Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, et al. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014;12:87.
Simpson EH. Measurement of Diversity. Nature. 1949;163:688–688.
Shannon CE. A mathematical theory of communication. SIGMOBILE Mob Comput Commun Rev. 2001;5:3–55.
CHAO A, YANG MCK. Stopping rules and estimation for recapture debugging with unequal failure rates. Biometrika. 1993;80:193–201.
Colwell RK, Mao CX, Chang J. Interpolating, extrapolating, and comparing incidence-based species Accumulation curves. Ecology. 2004;85:2717–27.
Pielou EC. The measurement of diversity in different types of biological collections. J Theor Biol. 1966;13:131–44.
Bray JR, Curtis JT. An ordination of the Upland Forest Communities of Southern Wisconsin. Ecol Monogr. 1957;27:325–49.
Hefner R, Warren S, Torgerson. Theory and methods of scaling. New York: John Wiley and Sons, Inc., 1958. Pp. 460. Behav Sci. 1959;4:245–7.
Kurki MI, Karjalainen J, Palta P, Sipilä TP, Kristiansson K, Donner KM, et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature. 2023;613:508–18.
Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet. 2022;54:134–42.
Abeshouse A, Ahn J, Akbani R, Ally A, Amin S, Andry CD, et al. The Molecular Taxonomy of primary prostate Cancer. Cell. 2015;163:1011–25.
Parte AC, Sardà Carbasse J, Meier-Kolthoff JP, Reimer LC, Göker M. List of prokaryotic names with standing in nomenclature (LPSN) moves to the DSMZ. Int J Syst Evol Microbiol. 2020;70:5607–12.
Rizzo A, Santoni M, Mollica V, Fiorentino M, Brandi G, Massari F. Microbiota and prostate cancer. Semin Cancer Biol. 2022;86:1058–65.
COHEN RJ, SHANNON BA, McNEAL JE, SHANNON T, GARRETT KL. PROPIONIBACTERIUM ACNES ASSOCIATED WITH INFLAMMATION, IN RADICAL PROSTATECTOMY SPECIMENS: A POSSIBLE LINK TO CANCER EVOLUTION? J Urol [Internet]. 2005 [cited 2024 Aug 10]; https://auajournals.publicaciones.saludcastillayleon.es/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1097/01.ju.0000158161.15277.78
Sfanos KS, Sauvageot J, Fedor HL, Dick JD, De Marzo AM, Isaacs WB. A molecular analysis of prokaryotic and viral DNA sequences in prostate tissue from patients with prostate cancer indicates the presence of multiple and diverse microorganisms. Prostate. 2008;68:306–20.
Xu H, Leng J, Liu F, Chen T, Qu J, Yang Y, et al. Tumor microbiota of renal cell carcinoma affects clinical prognosis by influencing the tumor immune microenvironment. Heliyon. 2024;10:e38310.
Mouradov D, Greenfield P, Li S, In E-J, Storey C, Sakthianandeswaren A, et al. Oncomicrobial Community Profiling identifies clinicomolecular and prognostic subtypes of Colorectal Cancer. Gastroenterology. 2023;165:104–20.
Peters BA, Hayes RB, Goparaju C, Reid C, Pass HI, Ahn J. The Microbiome in Lung Cancer tissue and recurrence-free survival. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol. 2019;28:731–40.
Sfanos KS, Yegnasubramanian S, Nelson WG, De Marzo AM. The inflammatory microenvironment and microbiome in prostate cancer development. Nat Rev Urol. 2018;15:11–24.
Bui N-N, Li C-Y, Wang L-Y, Chen Y-A, Kao W-H, Chou L-F, et al. Clostridium scindens metabolites trigger prostate cancer progression through androgen receptor signaling. J Microbiol Immunol Infect Wei Mian Yu Gan Ran Za Zhi. 2023;56:246–56.
Liss MA, White JR, Goros M, Gelfond J, Leach R, Johnson-Pais T, et al. Metabolic biosynthesis pathways identified from fecal Microbiome Associated with prostate Cancer. Eur Urol. 2018;74:575.
Harris WP, Mostaghel EA, Nelson PS, Montgomery B. Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion. Nat Clin Pract Urol. 2009;6:76–85.
Zhang X, Cheng L, Gao C, Chen J, Liao S, Zheng Y, et al. Androgen Signaling contributes to sex differences in Cancer by inhibiting NF-κB activation in T cells and suppressing Antitumor Immunity. Cancer Res. 2023;83:906–21.
Dong J, Li Y, Xiao H, Zhang S, Wang B, Wang H, et al. Oral microbiota affects the efficacy and prognosis of radiotherapy for colorectal cancer in mouse models. Cell Rep. 2021;37:109886.
Lu Y-Q, Qiao H, Tan X-R, Liu N. Broadening oncological boundaries: the intratumoral microbiota. Trends Microbiol. 2024;32:807–22.
Lee J, Yang W, Hostetler A, Schultz N, Suckow MA, Stewart KL, et al. Characterization of the anti-inflammatory Lactobacillus reuteri BM36301 and its probiotic benefits on aged mice. BMC Microbiol. 2016;16:69.
Markle JGM, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, et al. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013;339:1084–8.
Acknowledgements
We thank the developers of the ‘Kraken2’ and R package ‘TwoSampleMR’.
We thank Mingjie Chen (Shanghai NewCore Biotechnology Co., Ltd.) for providing data analysis and visualization support.
Funding
This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.
Author information
Authors and Affiliations
Contributions
Y.G.C contribute to the article searching and data acquisition. Y.G.C and L.C.Z contribute to data cleaning. Y.G.C, P.H, X.J.C, M.L.T contribute to data analysis. Y.G.C drafted this manuscript. X.Q.D and W.S.G revised the manuscript. All authors contributed to the article and approved the submitted version. Y.G.C, X.Q.D, and W.S.G contributed equally to this work.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have 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.
12967_2024_5937_MOESM1_ESM.xlsx
Supplementary Material 1: The subgroup LEfSe analysis of (A) PSA value ( < = 0.2 ng/ml and > 0.2 ng/ml) (B) Tumor pathology stage (T1/T2 and T3/T4).
12967_2024_5937_MOESM3_ESM.jpg
Supplementary Material 3: The LEfSe analysis of microbiota in metastasis prostate cancer grouped by treatment not adjusted by FDR. A) Anti-androgen therapy; B) Chemotherapy; C) Imunnotherapy; D) Target therapy; E) Nuclear therapy.
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/.
About this article
Cite this article
Ye, GC., Peng, H., Xiang, JC. et al. Comprehensive analysis of the interaction microbiome and prostate cancer: an initial exploration from multi-cohort metagenome and GWAS studies. J Transl Med 23, 130 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05937-7
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05937-7