1
|
Arnone AA, Tsai YT, Cline JM, Wilson AS, Westwood B, Seger ME, Chiba A, Howard-McNatt M, Levine EA, Thomas A, Soto-Pantoja DR, Cook KL. Endocrine-targeting therapies shift the breast microbiome to reduce estrogen receptor-α breast cancer risk. Cell Rep Med 2024:101880. [PMID: 39742868 DOI: 10.1016/j.xcrm.2024.101880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/14/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Studies indicate that breast tissue has a distinct modifiable microbiome population. We demonstrate that endocrine-targeting therapies, such as tamoxifen, reshape the non-cancerous breast microbiome to influence tissue metabolism and reduce tumorigenesis. Using 16S sequencing, we found that tamoxifen alters β-diversity and increases Firmicutes abundance, including Lactobacillus spp., in mammary glands (MGs) of mice and non-human primates. Immunohistochemistry showed that lipoteichoic acid (LTA)-positive bacteria were elevated in tamoxifen-treated breast tissue. In B6.MMTV-PyMT mice, intra-nipple probiotic bacteria injections reduced tumorigenesis, altered metabolic gene expression, and decreased tumor proliferation. Probiotic-conditioned media selectively reduced viability in estrogen receptor-positive (ER+) breast cancer cells and altered mitochondrial metabolism in non-cancerous epithelial cells. Human tumor samples revealed that LTA-positive bacteria negatively correlated with Ki67, suggesting that endocrine therapies influence tumor-associated microbiota to regulate proliferation. Our data indicate that endocrine-targeting therapies modify the breast microbiome, corresponding with a shift in tissue metabolism to potentially reduce ER+ breast cancer risk.
Collapse
Affiliation(s)
- Alana A Arnone
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Yu-Ting Tsai
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian Westwood
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Meghan E Seger
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Akiko Chiba
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Durham VA Medical Center, Department of Surgery, Durham, NC 27705, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Marissa Howard-McNatt
- Department of General Surgery, Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem NC, 27157, USA
| | - Edward A Levine
- Department of General Surgery, Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem NC, 27157, USA
| | - Alexandra Thomas
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
2
|
Zhao H, Zhang L, Du D, Mai L, Liu Y, Morigen M, Fan L. The RIG-I-like receptor signaling pathway triggered by Staphylococcus aureus promotes breast cancer metastasis. Int Immunopharmacol 2024; 142:113195. [PMID: 39303544 DOI: 10.1016/j.intimp.2024.113195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Host microbes are increasingly recognized as key components in various types of cancer, although their exact impact remains unclear. This study investigated the functional significance of Staphylococcus aureus (S. aureus) in breast cancer tumorigenesis and progression. We found that S. aureus invasion resulted in a compromised DNA damage response process, as evidenced by the absence of G1-phase arrest and apoptosis in breast cells in the background of double strand breaks production and the activation of the ataxia-telangiectasia mutated (ATM)-p53 signaling pathway. The high-throughput mRNA sequencing, bioinformatics analysis and pharmacological studies revealed that S. aureus facilitates breast cell metastasis through the innate immune pathway, particularly in cancer cells. During metastasis, S. aureus initially induced the expression of RIG-I-like receptors (RIG-I in normal breast cells, RIG-I and MDA5 in breast cancer cells), which in turn activated NF-κB p65 expression. We further showed that NF-κB p65 activated the CCL5-CCR5 pathway, contributing to breast cell metastasis. Our study provides novel evidence that the innate immune system, triggered by bacterial infection, plays a role in bacterial-driven cancer metastasis.
Collapse
Affiliation(s)
- Haile Zhao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Linzhe Zhang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Dongdong Du
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Lisu Mai
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Yaping Liu
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Morigen Morigen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China.
| | - Lifei Fan
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China.
| |
Collapse
|
3
|
Ma Y, Chen T, Sun T, Dilimulati D, Xiao Y. The oncomicrobiome: New insights into microorganisms in cancer. Microb Pathog 2024; 197:107091. [PMID: 39481695 DOI: 10.1016/j.micpath.2024.107091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The discoveries of the oncomicrobiome (intratumoral microbiome) and oncomicrobiota (intratumoral microbiota) represent significant advances in tumor research and have rapidly become of key interest to the field. Within tumors, microorganisms such as bacteria, fungi, viruses, and archaea form the oncomicrobiota and are primarily found within tumor cells, immunocytes, and the intercellular matrix. The oncomicrobiome exhibits marked heterogeneity and is associated with tumor initiation, progression, metastasis, and treatment response. Interactions between the oncomicrobiome and the immune system can modulate host antitumor immunity, influencing the efficacy of immunotherapies. Oncomicrobiome research also faces numerous challenges, including overcoming methodological issues such as low target abundance, susceptibility to contamination, and biases in sample handling and analysis methods across different studies. Furthermore, studies of the oncomicrobiome may be confounded by baseline differences in microbiomes among populations driven by both environmental and genetic factors. Most studies to date have revealed associations between the oncomicrobiome and tumors, but very few have established mechanistic links between the two. This review introduces the relevant concepts, detection methods, sources, and characteristics of the oncomicrobiome. We then describe the composition of the oncomicrobiome in common tumors and its role in shaping the tumor microenvironment. We also discuss the current problems and challenges to be overcome in this rapidly progressing field.
Collapse
Affiliation(s)
- Yingying Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingting Sun
- Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China; Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Dilinuer Dilimulati
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China; Peking Union Medical College & Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Cui J, Li X, Zhang Q, Du B, Ding Z, Yan C, Xue G, Gan L, Feng J, Fan Z, Xu Z, Yu Z, Fu T, Feng Y, Zhao H, Kong Y, Cui X, Tian Z, Liu Q, Yuan J. Existence and distribution of the microbiome in tumour tissues of children with hepatoblastoma. Heliyon 2024; 10:e39547. [PMID: 39553581 PMCID: PMC11564952 DOI: 10.1016/j.heliyon.2024.e39547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/18/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Cancer microbiota have recently been demonstrated in several cancer types. The microbiome enhances inflammation in the cancer microenvironment and affects the disease pathology by regulating tumourigenesis, cancer progression, and chemotherapy resistance. Hepatoblastoma (HB), the most common childhood malignant tumour, is a malignant embryonic tumour. However, the pathogenesis and molecular basis of HB remain poorly understood. In this study, to explore the existence and distribution of the microbiome in tumour tissues and adjacent non-tumour tissues of children with HB, we mainly performed 16S rDNA sequencing, and the results showed that the diversity and abundance of the microbiome in children with HB were significantly different between HB tumours and adjacent non-tumour tissues (p < 0.01). At the phylum level, the dominant microbiome in the tumour tissues were Proteobacteria, Bacteroidetes, and Firmicutes. At the genus level, Ruminococcus was more abundant in HB tumours than in the adjacent non-tumour tissues. Simultaneously, the abundances of Bacteroides, Parabacteroides, Lachnospiracea-NK4A136, and Alistipes in HB tumours were lower than those in the adjacent non-tumour tissues. In addition, Romboutsia strongly correlated with alpha-fetoprotein, an important indicator of HB. Sphingomonas was abundant in primary HB tumours, whereas Oscillibacter and Pandoraea were abundant in metastatic HB tumours. However, whether these bacteria are associated with HB needs further evaluation. Therefore, we identified the microbiome that correlated with the occurrence and development of HB. Ruminococcus and Romboutsia were identified as potential bacterial markers of HB tumours. To conclude, we found that HB also contains cancer microbiome, and it is necessary to shed light on the microbiome characteristics of HB in the future.
Collapse
Affiliation(s)
- Jinghua Cui
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Xiaoran Li
- Postgraduate Base of the PLA Rocket Force Medical Center, Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
- Department of Hematology and Oncology, 155th Hospital of Kaifeng, Kaifeng, 475003, Henan Province, China
| | - Qun Zhang
- Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Bing Du
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zanbo Ding
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Chao Yan
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Guanhua Xue
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lin Gan
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Junxia Feng
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zheng Fan
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziying Xu
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zihui Yu
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Tongtong Fu
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yanling Feng
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hanqing Zhao
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yiming Kong
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Xiaohu Cui
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziyan Tian
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Quanda Liu
- Postgraduate Base of the PLA Rocket Force Medical Center, Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
- Department of General Surgery, Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jing Yuan
- Capital Institute of Pediatrics, Beijing, 100020, China
| |
Collapse
|
5
|
Rozani S, Lykoudis PM. The impact of intestinal and mammary microbiomes on breast cancer development: A review on the microbiota and oestrobolome roles in tumour microenvironments. Am J Surg 2024; 237:115795. [PMID: 38853033 DOI: 10.1016/j.amjsurg.2024.115795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Microbiota affects carcinogenesis by altering energy equilibrium, increasing fat mass, synthesizing small signaling molecules, and formulating and regulating immune response and indigestible food ingredient, xenobiotic, and pharmaceutical compound metabolism. The intestinal microbiome can moderate oestrogen and other steroid hormone metabolisms, and secrete bioactive metabolites that are important for tumour microenvironment. Specifically, the breast tissue microbiome could become altered and lead to breast cancer development. The study of oestrobolome, the microbiomic component that metabolizes oestrogens, can contribute to better breast cancer understanding and subsequent treatment. Investigating oestrobolome-related oestrogen metabolism mechanisms in immune system regulation can shed light on how intestinal microorganisms regulate tumour microenvironment. Intestinal and regional breast microbiomes can determine treatment lines and serve as possible biomarkers for breast cancer. The aim of this study is to summarise current evidence on the role of microbiome in breast cancer progression with particular interest in therapeutic and diagnostic implementation.
Collapse
Affiliation(s)
- Sofia Rozani
- Faculty of Medicine, National and Kapodistrian University of Athens, Greece.
| | - Panagis M Lykoudis
- Faculty of Medicine, National and Kapodistrian University of Athens, Greece; Honorary Lecturer, Division of Surgery and Interventional Science, University College London (UCL), United Kingdom
| |
Collapse
|
6
|
Liu MM, Zhu HH, Bai J, Tian ZY, Zhao YJ, Boekhout T, Wang QM. Breast cancer colonization by Malassezia globosa accelerates tumor growth. mBio 2024; 15:e0199324. [PMID: 39235230 PMCID: PMC11481877 DOI: 10.1128/mbio.01993-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 09/06/2024] Open
Abstract
Malassezia globosa is a lipophilic basidiomycetous yeast that occurs abundantly in breast tumors and that may contribute to a shortened overall survival of breast cancer (BRAC) patients, suggesting that the yeast may participate in the carcinogenesis of BRAC. However, the mechanisms involved in the M. globosa-based acceleration of BRAC are unknown. Here, we show that M. globosa can colonize mammary tissue in 7,12-dimethylbenz[a] anthracene-induced mice. The abundance of M. globosa shortened the overall survival and increased the tumor incidence. Transcriptome data illustrated that IL-17A plays a key role in tumor growth due to M. globosa colonization, and tumor-associated macrophage infiltration was elevated during M. globosa colonization which triggers M2 polarization of macrophages via toll-like receptors 4/nuclear factor kappa-B (Nf-κB) signaling. Our results show that the expression of sphingosine kinase 1 (Sphk1) is increased in breast tumors after inoculation with M. globosa. Moreover, we discovered that Sphk1-specific small interfering RNA blocked the formation of lipid droplets, which can effectively alleviate the expression of the signal transducer and activator of the transcription 3 (STAT3)/Nf-κB pathway. Taken together, our results demonstrate that M. globosa could be a possible factor for the progression of BRAC. The mechanisms by which M. globosa promotes BRAC development involve the IL-17A/macrophage axis. Meanwhile, Sphk1 overexpression was induced by M. globosa infection, which also promoted the proliferation of MCF-7 cells.IMPORTANCELiterature has suggested that Malassezia globosa is associated with breast tumors; however, this association has not been confirmed. Here, we found that M. globosa colonizes in breast fat pads leading to tumor growth. As a lipophilic yeast, the expression of sphingosine kinase 1 (Sphk1) was upregulated to promote tumor growth after M. globosa colonization. Moreover, the IL-17A/macrophages axis plays a key role in mechanisms involved in the M. globosa-induced breast cancer acceleration from the tumor immune microenvironment perspective.
Collapse
Affiliation(s)
- Miao-Miao Liu
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, Hebei, China
| | - Hui-Hui Zhu
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, Hebei, China
| | - Jie Bai
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, Hebei, China
| | - Zi-Ye Tian
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, Hebei, China
| | - Yu-Jing Zhao
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, Hebei, China
| | - Teun Boekhout
- College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Qi-Ming Wang
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, Hebei, China
- Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding, Hebei, China
- Engineering Research Center of Ecological Safety and Conservation in Beijing-Tianjin-Hebei (Xiong’an New Area) of MOE, Xiong’an, China
| |
Collapse
|
7
|
Moreno‐Arrones OM, Béa‐Ardebol S, Galiano‐Mejías S, Turrión‐Merino L, de Perosanz‐Lobo D, Perez‐Brocal V, Moya A, Rios‐Buceta L. Locally advanced basal cell carcinoma associated with distinct gut microbiome signature. J Dtsch Dermatol Ges 2024. [DOI: 10.1111/ddg.15588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 09/04/2024] [Indexed: 12/03/2024]
Affiliation(s)
- Oscar M. Moreno‐Arrones
- Dermatology Department Ramon y Cajal University Hospital University of Alcala Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Clínica Pedro Jaén Madrid Spain
| | - Sonia Béa‐Ardebol
- Dermatology Department Ramon y Cajal University Hospital University of Alcala Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Clínica Pedro Jaén Madrid Spain
| | | | | | - Dario de Perosanz‐Lobo
- Dermatology Department Ramon y Cajal University Hospital University of Alcala Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Clínica Pedro Jaén Madrid Spain
| | - Vincente Perez‐Brocal
- Department of Genomics and Health Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO‐Public Health) Valencia Spain
- CIBER in Epidemiology and Public Health (CIBEResp) Madrid Spain
| | - Andres Moya
- Department of Genomics and Health Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO‐Public Health) Valencia Spain
- CIBER in Epidemiology and Public Health (CIBEResp) Madrid Spain
- Institute for Integrative Systems Biology (I2SysBio) The University of Valencia and The Spanish National Research Council (CSIC)‐UVEG) Valencia Spain
| | - Luis Rios‐Buceta
- Dermatology Department Ramon y Cajal University Hospital University of Alcala Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Clínica Pedro Jaén Madrid Spain
| |
Collapse
|
8
|
Neagoe CXR, Ionică M, Neagoe OC, Trifa AP. The Influence of Microbiota on Breast Cancer: A Review. Cancers (Basel) 2024; 16:3468. [PMID: 39456562 PMCID: PMC11506631 DOI: 10.3390/cancers16203468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Breast cancer remains one of the leading causes of death among women worldwide, and recent research highlights its growing connection to alterations in the microbiota. This review delves into the intricate relationship between microbiotas and breast cancer, exploring its presence in healthy breast tissue, its changes during cancer progression, and its considerable impact on both the tumor microenvironment (TME) and the tumor immune microenvironment (TIME). We extensively analyze how the microbiota influences cancer growth, invasion, metastasis, resistance to drugs, and the evasion of the immune system, with a special focus on its effects on the TIME. Furthermore, we investigate distinct microbial profiles associated with the four primary molecular subtypes of breast cancer, examining how the microbiota in tumor tissues compares with that in adjacent normal tissues. Emerging studies suggest that microbiotas could serve as valuable diagnostic and prognostic biomarkers, as well as targets for therapy. This review emphasizes the urgent need for further research to improve strategies for breast cancer prevention, diagnosis, and treatment. By offering a detailed examination of the microbiota's critical role in breast cancer, this review aims to foster the development of novel microbiota-based approaches for managing the disease.
Collapse
Affiliation(s)
- Cara-Xenia-Rafaela Neagoe
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Mihaela Ionică
- Second Clinic of General Surgery and Surgical Oncology, Emergency Clinical Municipal Hospital, 300079 Timișoara, Romania;
- Second Discipline of Surgical Semiology, First Department of Surgery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Breast Surgery Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300079 Timișoara, Romania
| | - Octavian Constantin Neagoe
- Second Clinic of General Surgery and Surgical Oncology, Emergency Clinical Municipal Hospital, 300079 Timișoara, Romania;
- Second Discipline of Surgical Semiology, First Department of Surgery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Breast Surgery Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300079 Timișoara, Romania
| | - Adrian Pavel Trifa
- The Discipline of Genetics, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Genetics, Clinical Hospital of Infectious Diseases and Pneumophthisiology “Dr. Victor Babes” Timisoara, 300041 Timisoara, Romania
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
9
|
Mikó E, Sipos A, Tóth E, Lehoczki A, Fekete M, Sebő É, Kardos G, Bai P. Guideline for designing microbiome studies in neoplastic diseases. GeroScience 2024; 46:4037-4057. [PMID: 38922379 PMCID: PMC11336004 DOI: 10.1007/s11357-024-01255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Oncobiosis has emerged as a key contributor to the development, and modulator of the treatment efficacy of cancer. Hereby, we review the modalities through which the oncobiome can support the progression of tumors, and the emerging therapeutic opportunities they present. The review highlights the inherent challenges and limitations faced in sampling and accurately characterizing oncobiome. Additionally, the review underscores the critical need for the standardization of microbial analysis techniques and the consistent reporting of microbiome data. We provide a suggested metadata set that should accompany microbiome datasets from oncological settings so that studies remain comparable and decipherable.
Collapse
Affiliation(s)
- Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary
| | - Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group, 4032, Debrecen, Hungary
| | - Andrea Lehoczki
- Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital-National Institute for Hematology and Infectious Diseases, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Éva Sebő
- Breast Center, Kenézy Gyula Hospital, University of Debrecen, 4032, Debrecen, Hungary
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, 4032, Debrecen, Hungary
- Faculty of Health Sciences, One Health Institute, University of Debrecen, 4032, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary.
- HUN-REN-DE Cell Biology and Signaling Research Group, 4032, Debrecen, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032, Debrecen, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
- Center of Excellence, The Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
10
|
Li N, Xiao C, Li Y, Zhang Y, Lin Y, Liu Q, Tang L, Xu L, Ren Z. Association of Chlamydia trachomatis Infection With Breast Cancer Risk and the Modification Effect of IL-12. Clin Breast Cancer 2024; 24:e554-e559.e1. [PMID: 38821744 DOI: 10.1016/j.clbc.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/07/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Chlamydia trachomatis (C. trachomatis) infection has been implicated in various cancers, yet its association with breast cancer remains unexplored. This infection triggers a cascade of immune responses primarily regulated by Interleukins-12 (IL-12). Thus, the objective of this case-control study was to investigate the link between C. trachomatis infection and breast cancer risk, as well as the modification effect of IL-12. METHODS We assessed IgG levels against C. trachomatis in serum of 1,121 women with breast cancer (861 with estrogen receptor-positive (ER+) and 260 with estrogen receptor-negative (ER-) tumors) and 400 controls in Guangzhou, China. Logistic regression models were applied to estimate the odds ratios (ORs) and 95% confidence intervals (95% CIs) for breast cancer risk in association with C. trachomatis infection. The interaction between C. trachomatis infection and IL-12 on breast cancer risk was estimated by the product terms in the logistic regression models. RESULTS Seropositivity of C. trachomatis IgG showed a slight association with an increased risk of breast cancer (OR = 1.20; 95% CI: 0.86∼1.78). This association was more pronounced among women with a higher (OR = 5.82; 95% CI: 1.31∼25.94) than a lower (OR = 0.73; 95% CI: 0.41∼1.30) level of IL-12, with a statistically significant interaction observed (Pinteraction = 0.013). In addition, C. trachomatis IgG seropositivity was related to an increased risk of breast cancer among PR+ patients (OR = 1.53; 95% CI: 1.04∼2.23). CONCLUSIONS C. trachomatis infection may contribute to the development of hormone-responsive breast cancer in women with high levels of IL-12. Further studies are needed to uncover the underlying mechanisms.
Collapse
Affiliation(s)
- Na Li
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Chengkun Xiao
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yunqian Li
- School of Public Health, Sun Yat-sen University, Guangzhou, China; Shenzhen Pingle Orthopedic Hospital, Shenzhen, China
| | - Yixin Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiang Liu
- Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Luying Tang
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, Guangzhou, China; School of Public Health, The University of Hong Kong, Hong Kong; Institute of Applied Health Research, University of Birmingham, Birmingham, UK.
| | - Zefang Ren
- School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
11
|
Le Ngoc K, Pham TTH, Nguyen TK, Huong PT. Pharmacomicrobiomics in precision cancer therapy: bench to bedside. Front Immunol 2024; 15:1428420. [PMID: 39315107 PMCID: PMC11416994 DOI: 10.3389/fimmu.2024.1428420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The burgeoning field of pharmacomicrobiomics offers promising insights into the intricate interplay between the microbiome and cancer, shaping responses to diverse treatment modalities. This review aims to analyze the molecular mechanisms underlying interactions between distinct microbiota types and cancer, as well as their influence on treatment outcomes. We explore how the microbiome impacts antitumor immunity, and response to chemotherapy, immunotherapy, and radiation therapy, unveiling its multifaceted roles in cancer progression and therapy resistance. Moreover, we discuss the challenges hindering the development of microbiome-based interventions in cancer therapy, including standardization, validation, and clinical translation. By synthesizing clinical evidence, we underscore the transformative potential of harnessing pharmacomicrobiomics in guiding cancer treatment decisions, paving the way for improved patient outcomes in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Phung Thanh Huong
- Faculty of Biotechnology, Hanoi University of Pharmacy,
Hanoi, Vietnam
| |
Collapse
|
12
|
Sheng D, Jin C, Yue K, Yue M, Liang Y, Xue X, Li P, Zhao G, Zhang L. Pan-cancer atlas of tumor-resident microbiome, immunity and prognosis. Cancer Lett 2024; 598:217077. [PMID: 38908541 DOI: 10.1016/j.canlet.2024.217077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/23/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
The existence of microbiome in human tumors has been determined widely, but evaluating the contribution of intratumoral bacteria and fungi to tumor immunity and prognosis from a pan-cancer perspective remains absent. We designed an improved microbial analysis pipeline to reduce interference from host sequences, complemented with integration analysis of intratumoral microbiota at species level with clinical indicators, tumor microenvironment, and prognosis across cancer types. We found that intratumoral microbiota is associated with immunophenotyping, with high-immunity subtypes showing greater bacterial and fungal richness compared to low-immunity groups. We also noted that the combination of fungi and bacteria demonstrated promising prognostic value across cancer types. We, thus, present The Cancer Microbiota (TCMbio), an interactive platform that provides the intratumoral bacteria and fungi data, and a comprehensive analysis module for 33 types of cancers. This led to the discovery of clinical and prognostic significance of intratumoral microbes.
Collapse
Affiliation(s)
- Dashuang Sheng
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Chuandi Jin
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kaile Yue
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Yue
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yijia Liang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinxin Xue
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pingfu Li
- Shandong Huxley Medical Technology Co.,Ltd., Jinan, China
| | - Guoping Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China; CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.
| |
Collapse
|
13
|
Naik A, Godbole M. Elucidating the Intricate Roles of Gut and Breast Microbiomes in Breast Cancer Metastasis to the Bone. Cancer Rep (Hoboken) 2024; 7:e70005. [PMID: 39188104 PMCID: PMC11347752 DOI: 10.1002/cnr2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 07/10/2024] [Accepted: 08/11/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Breast cancer is the most predominant and heterogeneous cancer in women. Moreover, breast cancer has a high prevalence to metastasize to distant organs, such as the brain, lungs, and bones. Patients with breast cancer metastasis to the bones have poor overall and relapse-free survival. Moreover, treatment using chemotherapy and immunotherapy is ineffective in preventing or reducing cancer metastasis. RECENT FINDINGS Microorganisms residing in the gut and breast, termed as the resident microbiome, have a significant influence on the formation and progression of breast cancer. Recent studies have identified some microorganisms that induce breast cancer metastasis to the bone. These organisms utilize multiple mechanisms, including induction of epithelial-mesenchymal transition, steroid hormone metabolism, immune modification, bone remodeling, and secretion of microbial products that alter tumor microenvironment, and enhance propensity of breast cancer cells to metastasize. However, their involvement makes these microorganisms suitable as novel therapeutic targets. Thus, studies are underway to prevent and reduce breast cancer metastasis to distant organs, including the bone, using chemotherapeutic or immunotherapeutic drugs, along with probiotics, antibiotics or fecal microbiota transplantation. CONCLUSIONS The present review describes association of gut and breast microbiomes with bone metastases. We have elaborated on the mechanisms utilized by breast and gut microbiomes that induce breast cancer metastasis, especially to the bone. The review also highlights the current treatment options that may target both the microbiomes for preventing or reducing breast cancer metastases. Finally, we have specified the necessity of maintaining a diverse gut microbiome to prevent dysbiosis, which otherwise may induce breast carcinogenesis and metastasis especially to the bone. The review may facilitate more detailed investigations of the causal associations between these microbiomes and bone metastases. Moreover, the potential treatment options described in the review may promote discussions and research on the modes to improve survival of patients with breast cancer by targeting the gut and breast microbiomes.
Collapse
Affiliation(s)
- Amruta Naik
- Department of Biosciences and Technology, School of Science and Environmental StudiesDr. Vishwanath Karad MIT World Peace UniversityPuneIndia
| | - Mukul S. Godbole
- Department of Biosciences and Technology, School of Science and Environmental StudiesDr. Vishwanath Karad MIT World Peace UniversityPuneIndia
| |
Collapse
|
14
|
Maranha A, Alarico S, Nunes-Costa D, Melo-Marques I, Roxo I, Castanheira P, Caramelo O, Empadinhas N. Drinking Water Microbiota, Entero-Mammary Pathways, and Breast Cancer: Focus on Nontuberculous Mycobacteria. Microorganisms 2024; 12:1425. [PMID: 39065193 PMCID: PMC11279143 DOI: 10.3390/microorganisms12071425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The prospect of drinking water serving as a conduit for gut bacteria, artificially selected by disinfection strategies and a lack of monitoring at the point of use, is concerning. Certain opportunistic pathogens, notably some nontuberculous mycobacteria (NTM), often exceed coliform bacteria levels in drinking water, posing safety risks. NTM and other microbiota resist chlorination and thrive in plumbing systems. When inhaled, opportunistic NTM can infect the lungs of immunocompromised or chronically ill patients and the elderly, primarily postmenopausal women. When ingested with drinking water, NTM often survive stomach acidity, reach the intestines, and migrate to other organs using immune cells as vehicles, potentially colonizing tumor tissue, including in breast cancer. The link between the microbiome and cancer is not new, yet the recognition of intratumoral microbiomes is a recent development. Breast cancer risk rises with age, and NTM infections have emerged as a concern among breast cancer patients. In addition to studies hinting at a potential association between chronic NTM infections and lung cancer, NTM have also been detected in breast tumors at levels higher than normal adjacent tissue. Evaluating the risks of continued ingestion of contaminated drinking water is paramount, especially given the ability of various bacteria to migrate from the gut to breast tissue via entero-mammary pathways. This underscores a pressing need to revise water safety monitoring guidelines and delve into hormonal factors, including addressing the disproportionate impact of NTM infections and breast cancer on women and examining the potential health risks posed by the cryptic and unchecked microbiota from drinking water.
Collapse
Affiliation(s)
- Ana Maranha
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Susana Alarico
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniela Nunes-Costa
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Inês Melo-Marques
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Inês Roxo
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Olga Caramelo
- Gynecology Department, Coimbra Hospital and University Centre (CHUC), 3004-561 Coimbra, Portugal;
| | - Nuno Empadinhas
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
15
|
Amaro-da-Cruz A, Rubio-Tomás T, Álvarez-Mercado AI. Specific microbiome patterns and their association with breast cancer: the intestinal microbiota as a potential biomarker and therapeutic strategy. Clin Transl Oncol 2024:10.1007/s12094-024-03554-w. [PMID: 38890244 DOI: 10.1007/s12094-024-03554-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer (BC) is one of the most diagnosed cancers in women. Based on histological characteristics, they are classified as non-invasive, or in situ (tumors located within the milk ducts or milk lobules) and invasive. BC may develop from in situ carcinomas over time. Determining prognosis and predicting response to treatment are essential tools to manage this disease and reduce its incidence and mortality, as well as to promote personalized therapy for patients. However, over half of the cases are not associated with known risk factors. In addition, some patients develop resistance to treatment and relapse. Therefore, it is necessary to identify new biomarkers and treatment strategies that improve existing therapies. In this regard, the role of the microbiome is being researched as it could play a role in carcinogenesis and the efficacy of BC therapies. This review aims to describe specific microbiome patterns associated with BC. For this, a literature search was carried out in PubMed database using the MeSH terms "Breast Neoplasms" and "Gastrointestinal Microbiome", including 29 publications. Most of the studies have focused on characterizing the gut or breast tissue microbiome of the patients. Likewise, studies in animal models and in vitro that investigated the impact of gut microbiota (GM) on BC treatments and the effects of the microbiome on tumor cells were included. Based on the results of the included articles, BC could be associated with an imbalance in the GM. This imbalance varied depending on molecular type, stage and grade of cancer, menopause, menarche, body mass index, and physical activity. However, a specific microbial profile could not be identified as a biomarker. On the other hand, some studies suggest that the GM may influence the efficacy of BC therapies. In addition, some microorganisms and bacterial metabolites could improve the effects of therapies or influence tumor development.
Collapse
Affiliation(s)
- Alba Amaro-da-Cruz
- Department of Chemical Engineering, Faculty of Science, University of Granada, 18071, Granada, Spain
| | - Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Ana I Álvarez-Mercado
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014, Granada, Spain.
- Institute of Nutrition and Food Technology, Biomedical Research Center, University of Granada, 18016, Armilla, Spain.
- Department of Pharmacology School of Pharmacy, University of Granada, 18071, Granada, Spain.
| |
Collapse
|
16
|
Wu-Chuang A, Mateos-Hernandez L, Abuin-Denis L, Maitre A, Avellanet J, García A, Fuentes D, Cabezas-Cruz A. Exploring the impact of breast cancer on colonization resistance of mouse microbiota using network node manipulation. Heliyon 2024; 10:e30914. [PMID: 38784541 PMCID: PMC11112314 DOI: 10.1016/j.heliyon.2024.e30914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Breast cancer, a global health concern affecting women, has been linked to alterations in the gut microbiota, impacting various aspects of human health. This study investigates the interplay between breast cancer and the gut microbiome, particularly focusing on colonization resistance-an essential feature of the microbiota's ability to prevent pathogenic overgrowth. Using a mouse model of breast cancer, we employ diversity analysis, co-occurrence network analysis, and robustness tests to elucidate the impact of breast cancer on microbiome dynamics. Our results reveal that breast cancer exposure affects the bacterial community's composition and structure, with temporal dynamics playing a role. Network analysis demonstrates that breast cancer disrupts microbial interactions and decreases network complexity, potentially compromising colonization resistance. Moreover, network robustness analysis shows the susceptibility of the microbiota to node removal, indicating potential vulnerability to pathogenic colonization. Additionally, predicted metabolic profiling of the microbiome highlights the significance of the enzyme EC 6.2.1.2 - Butyrate--CoA ligase, potentially increasing butyrate, and balancing the reduction of colonization resistance. The identification of Rubrobacter as a key contributor to this enzyme suggests its role in shaping the microbiota's response to breast cancer. This study uncovers the intricate relationship between breast cancer, the gut microbiome, and colonization resistance, providing insights into potential therapeutic strategies and diagnostic approaches for breast cancer patients.
Collapse
Affiliation(s)
- Alejandra Wu-Chuang
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | - Lourdes Mateos-Hernandez
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | - Lianet Abuin-Denis
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
- Animal Biotechnology Department, Center for Genetic Engineering and Biotechnology, Avenue 31 between 158 and 190, P.O. Box 6162, 10600, Havana, Cuba
| | - Apolline Maitre
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
- INRAE, UR 0045 Laboratoire de Recherches Sur Le Développement de L'Elevage (SELMET-LRDE), Corte, France
- EA 7310, Laboratoire de Virologie, Université de Corse, Corte, France
| | - Janet Avellanet
- Center of Molecular Immunology (CIM), Calle 15 esq. 216, Atabey, Playa, Havana, Cuba
| | - Arlem García
- Center of Molecular Immunology (CIM), Calle 15 esq. 216, Atabey, Playa, Havana, Cuba
| | - Dasha Fuentes
- National Center for Laboratory Animal Breeding (CENPALAB), Calle 3ra # 40759 entre 6ta y carretera de Tirabeque, Rpto La Unión, Boyeros, Havana, Cuba
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| |
Collapse
|
17
|
Patel NJ, Thippani S, Jathan J, Gaur G, Sawant JY, Pandya JM, Sapi E. Evidence for the presence of Borrelia burgdorferi in invasive breast cancer tissues. Eur J Microbiol Immunol (Bp) 2024; 14:143-153. [PMID: 38451280 PMCID: PMC11097788 DOI: 10.1556/1886.2024.00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Borrelia burgdorferi, the causative agent of Lyme disease, has recently been demonstrated to infect and enhance the invasive properties of breast cancer cells, while also influencing the expression of inflammatory chemokines (CXCL8 and CXCL10). This study investigates the presence of B. burgdorferi in invasive breast cancer tissues using commercially available, FDA-approved breast cancer tissue microarrays consisting of 350 ductal, 32 lobular, and 22 intraductal invasive breast carcinomas, alongside 29 normal breast tissues. Employing fluorescent immunohistochemical staining and high-resolution imaging, the findings revealed that approximately 20% of invasive lobular and ductal carcinomas, followed by 14% of intraductal carcinomas, tested positive for B. burgdorferi, while all normal breast tissues tested negative. PCR analysis further confirmed the presence of B. burgdorferi DNA in breast cancer tissues. Moreover, 25% of B. burgdorferi-positive tissues exhibited expression of both chemokines, CXCL8 and CXCL10, which was not observed in B. burgdorferi-negative tissues. Analysis of available patient data, including age, indicated a correlation between older patients and B. burgdorferi-positive tissues. This study validates the presence of B. burgdorferi in invasive breast cancer tissues and highlights the involvement of key CXCL family members associated with inflammatory processes.
Collapse
Affiliation(s)
- Niraj Jatin Patel
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Sahaja Thippani
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Jasmine Jathan
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Gauri Gaur
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Janhavi Y. Sawant
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Jay M. Pandya
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Eva Sapi
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| |
Collapse
|
18
|
Valsecchi AA, Ferrari G, Paratore C, Dionisio R, Vignani F, Sperone P, Vellani G, Novello S, Di Maio M. Gut and local microbiota in patients with cancer: increasing evidence and potential clinical applications. Crit Rev Oncol Hematol 2024; 197:104328. [PMID: 38490281 DOI: 10.1016/j.critrevonc.2024.104328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/19/2023] [Accepted: 03/11/2024] [Indexed: 03/17/2024] Open
Abstract
In recent years, cancer research has highlighted the role of disrupted microbiota in carcinogenesis and cancer recurrence. However, microbiota may also interfere with drug metabolism, influencing the efficacy of cancer drugs, especially immunotherapy, and modulating the onset of adverse events. Intestinal micro-organisms can be altered by external factors, such as use of antibiotics, proton pump inhibitors treatment, lifestyle and the use of prebiotics or probiotics. The aim of our review is to provide a picture of the current evidence about preclinical and clinical data of the role of gut and local microbiota in malignancies and its potential clinical role in cancer treatments. Standardization of microbiota sequencing approaches and its modulating strategies within prospective clinical trials could be intriguing for two aims: first, to provide novel potential biomarkers both for early cancer detection and for therapeutic effectiveness; second, to propose personalized and "microbiota-tailored" treatment strategies.
Collapse
Affiliation(s)
- Anna Amela Valsecchi
- Department of Oncology, University of Turin, Città della Salute e della Scienza di Torino, Turin, Italy
| | - Giorgia Ferrari
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Chiara Paratore
- Department of Oncology, ASL TO4, Ivrea Community Hospital, Ivrea, Italy.
| | - Rossana Dionisio
- Department of Oncology, University of Turin, Mauriziano Hospital, Turin, Italy
| | - Francesca Vignani
- Department of Oncology, University of Turin, Mauriziano Hospital, Turin, Italy
| | - Paola Sperone
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Giorgio Vellani
- Department of Oncology, ASL TO4, Ivrea Community Hospital, Ivrea, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Città della Salute e della Scienza di Torino, Turin, Italy
| |
Collapse
|
19
|
Cheng W, Li F, Gao Y, Yang R. Fungi and tumors: The role of fungi in tumorigenesis (Review). Int J Oncol 2024; 64:52. [PMID: 38551162 PMCID: PMC10997370 DOI: 10.3892/ijo.2024.5640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
Fungi inhabit different anatomic sites in the human body. Advances in omics analyses of host‑microbiome interactions have tremendously improved our understanding of the effects of fungi on human health and diseases such as tumors. Due to the significant enrichment of specific fungi in patients with malignant tumors, the associations between fungi and human cancer have attracted an increasing attention in recent years. Indeed, cancer type‑specific fungal profiles have been found in different tumor tissues. Importantly, fungi also influence tumorigenesis through multiple factors, such as host immunity and bioactive metabolites. Microbiome interactions, host factors and fungal genetic and epigenetic factors could be involved in fungal enrichment in tumor tissues and/or in the conversion from a commensal fungus to a pathogenic fungus. Exploration of the interactions of fungi with the bacterial microbiome and the host may enable them to be a target for cancer diagnosis and treatment. In the present review, the associations between fungi and human cancer, cancer type‑specific fungal profiles and the mechanisms by which fungi cause tumorigenesis were discussed. In addition, possible factors that can lead to the enrichment of fungi in tumor tissues and/or the conversion of commensal fungi to pathogenic fungi, as well as potential therapeutic and preventive strategies for tumors based on intratumoral fungi were summarized.
Collapse
Affiliation(s)
- Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, P.R. China
| | - Fan Li
- Department of Immunology, Nankai University School of Medicine, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, P.R. China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, P.R. China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, P.R. China
- State Key Laboratory of Medicinal Chemical Biology, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, P.R. China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, P.R. China
| |
Collapse
|
20
|
Li J, Zhang Y, Cai Y, Yao P, Jia Y, Wei X, Du C, Zhang S. Multi-omics analysis elucidates the relationship between intratumor microbiome and host immune heterogeneity in breast cancer. Microbiol Spectr 2024; 12:e0410423. [PMID: 38442004 PMCID: PMC10986513 DOI: 10.1128/spectrum.04104-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Research has indicated that intratumor microbiomes affect the occurrence, progression, and therapeutic response in many cancer types by influencing the immune system. We aim to evaluate the characteristics of immune-related intratumor microbiomes (IRIMs) in breast cancer (BC) and search for potential prognosis prediction factors and treatment targets. The clinical information, microbiome data, transcriptomics data of The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) patients were obtained from Kraken-TCGA-Raw-Data and TCGA portal. The core tumor-infiltrating immune cell was identified using univariate Cox regression analysis. Based on consensus clustering analysis, BC patients were categorized into two immune subtypes, referred to as immune-enriched and immune-deficient subtypes. The immune-enriched subtype, characterized by higher levels of immune infiltration of CD8+ T and macrophage M1 cells, demonstrated a more favorable prognosis. Furthermore, significant differences in alpha-diversity and beta-diversity were observed between the two immune subtypes, and the least discriminant analysis effect size method identified 33 types of IRIMs. An intratumor microbiome-based prognostic signature consisting of four prognostic IRIMs (Acidibacillus, Succinimonas, Lachnoclostridium, and Pseudogulbenkiania) was constructed using the Cox proportional-hazard model, and it had great prognostic value. The prognostic IRIMs were correlated with immune gene expression and the sensitivity of chemotherapy drugs, specifically tamoxifen and docetaxel. In conclusion, our research has successfully identified two distinct immune subtypes in BC, which exhibit contrasting prognoses and possess unique epigenetic and intratumor microbiomes. The critical IRIMs were correlated with prognosis, tumor-infiltrating immune cell abundance, and immunotherapeutic efficacy in BC. Consequently, this study has identified potential IRIMs as biomarkers, providing a novel therapeutic approach for treating BC.IMPORTANCERecent research has substantiated the presence of the intratumor microbiome in tumor immune microenvironment, which could influence tumor occurrence and progression, as well as provide new opportunities for cancer diagnosis and treatment. This study identified the critical immune-related intratumor microbiome (Acidibacillus, Succinimonas, Lachnoclostridium, and Pseudogulbenkiania), which were correlated with prognosis, tumor-infiltrating immune cell abundance, and immunotherapeutic efficacy in breast cancer and might be the novel target to regulate immunotherapy in BC.
Collapse
Affiliation(s)
- Jia Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Yu Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Yifan Cai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Peizhuo Yao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Yiwei Jia
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinyu Wei
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Chong Du
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Shuqun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
21
|
Li X, Xu H, Du Z, Cao Q, Liu X. Advances in the study of tertiary lymphoid structures in the immunotherapy of breast cancer. Front Oncol 2024; 14:1382701. [PMID: 38628669 PMCID: PMC11018917 DOI: 10.3389/fonc.2024.1382701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Breast cancer, as one of the most common malignancies in women, exhibits complex and heterogeneous pathological characteristics across different subtypes. Triple-negative breast cancer (TNBC) and HER2-positive breast cancer are two common and highly invasive subtypes within breast cancer. The stability of the breast microbiota is closely intertwined with the immune environment, and immunotherapy is a common approach for treating breast cancer.Tertiary lymphoid structures (TLSs), recently discovered immune cell aggregates surrounding breast cancer, resemble secondary lymphoid organs (SLOs) and are associated with the prognosis and survival of some breast cancer patients, offering new avenues for immunotherapy. Machine learning, as a form of artificial intelligence, has increasingly been used for detecting biomarkers and constructing tumor prognosis models. This article systematically reviews the latest research progress on TLSs in breast cancer and the application of machine learning in the detection of TLSs and the study of breast cancer prognosis. The insights provided contribute valuable perspectives for further exploring the biological differences among different subtypes of breast cancer and formulating personalized treatment strategies.
Collapse
Affiliation(s)
- Xin Li
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Han Xu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ziwei Du
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Cao
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, China
| | - Xiaofei Liu
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
22
|
Wu D, Guan YX, Li CH, Zheng Q, Yin ZJ, Wang H, Liu NN. "Nutrient-fungi-host" tripartite interaction in cancer progression. IMETA 2024; 3:e170. [PMID: 38882486 PMCID: PMC11170973 DOI: 10.1002/imt2.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 06/18/2024]
Abstract
The human microbiome exhibits a profound connection with the cancer development, progression, and therapeutic response, with particular emphasis on its components of the mycobiome, which are still in the early stages of research. In this review, we comprehensively summarize cancer-related symbiotic and pathogenic fungal genera. The intricate mechanisms through which fungi impact cancer as an integral member of both gut and tissue-resident microbiomes are further discussed. In addition, we shed light on the pivotal physiological roles of various nutrients, including cholesterol, carbohydrates, proteins and minerals, in facilitating the growth, reproduction, and invasive pathogenesis of the fungi. While our exploration of the interplay between nutrients and cancer, mediated by the mycobiome, is ongoing, the current findings have yet to yield conclusive results. Thus, delving into the relationship between nutrients and fungal pathogenesis in cancer development and progression would provide valuable insights into anticancer therapy and foster precision nutrition and individualized treatments that target fungi from bench to bedside.
Collapse
Affiliation(s)
- Di Wu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yun-Xuan Guan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Chen-Hao Li
- Institute of Computing Technology Chinese Academy of Sciences Beijing China
| | - Quan Zheng
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Zuo-Jing Yin
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
23
|
Thu MS, Pongpirul K, Vongsaisuwon M, Vinayanuwattikun C, Banchuen K, Ondee T, Payungporn S, Phutrakool P, Nootim P, Chariyavilaskul P, Cherdchom S, Wanaratna K, Hirankarn N. Efficacy and mechanisms of cannabis oil for alleviating side effects of breast cancer chemotherapy (CBC2): protocol for randomized controlled trial. BMC Complement Med Ther 2024; 24:130. [PMID: 38521934 PMCID: PMC10960413 DOI: 10.1186/s12906-024-04426-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND In a pilot study using both cannabidiol (CBD) and tetrahydrocannabinol (THC) as single agents in advanced cancer patients undergoing palliative care in Thailand, the doses were generally well tolerated, and the outcome measure of total symptom distress scores showed overall symptom benefit. The current study aims to determine the intensity of the symptoms experienced by breast cancer patients, to explore the microbiome profile, cytokines, and bacterial metabolites before and after the treatment with cannabis oil or no cannabis oil, and to study the pharmacokinetics parameters and pharmacogenetics profile of the doses. METHODS A randomized, double-blinded, placebo-controlled trial will be conducted on the breast cancer cases who were diagnosed with breast cancer and currently receiving chemotherapy at King Chulalongkorn Memorial Hospital (KCMH), Bangkok, Thailand. Block randomization will be used to allocate the patients into three groups: Ganja Oil (THC 2 mg/ml; THC 0.08 mg/drop, and CBD 0.02 mg/drop), Metta Osot (THC 81 mg/ml; THC 3 mg/drop), and placebo oil. The Edmonton Symptom Assessment System (ESAS), Food Frequency Questionnaires (FFQ), microbiome profile, cytokines, and bacterial metabolites will be assessed before and after the interventions, along with pharmacokinetic and pharmacogenetic profile of the treatment during the intervention. TRIAL REGISTRATION TCTR20220809001.
Collapse
Affiliation(s)
- May Soe Thu
- Joint Chulalongkorn University-University of Liverpool PhD Programme in Biomedical Sciences and Biotechnology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Infection Biology & Microbiomes, University of Liverpool, Liverpool, UK
| | - Krit Pongpirul
- Department of Infection Biology & Microbiomes, University of Liverpool, Liverpool, UK.
- Center of Excellence in Preventive and Integrative Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Bumrungrad International Hospital, Bangkok, Thailand.
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Mawin Vongsaisuwon
- Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chanida Vinayanuwattikun
- King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Department of Medicine, Faculty of Medicine, Division of Medical Oncology, Chulalongkorn University, Bangkok, Thailand
| | - Kamonwan Banchuen
- Center of Excellence in Preventive and Integrative Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Nonthaburi, Thailand
| | - Thunnicha Ondee
- Department of Infection Biology & Microbiomes, University of Liverpool, Liverpool, UK
| | - Sunchai Payungporn
- Department of Biochemistry, Faculty of Medicine, Center of Excellence in Systems Microbiology,, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Phanupong Phutrakool
- Center of Excellence in Preventive and Integrative Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chula Data Management Center, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Preecha Nootim
- Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Nonthaburi, Thailand
| | - Pajaree Chariyavilaskul
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sarocha Cherdchom
- Center of Excellence in Preventive and Integrative Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kulthanit Wanaratna
- Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Nonthaburi, Thailand
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
24
|
Hurst R, Brewer DS, Gihawi A, Wain J, Cooper CS. Cancer invasion and anaerobic bacteria: new insights into mechanisms. J Med Microbiol 2024; 73:001817. [PMID: 38535967 PMCID: PMC10995961 DOI: 10.1099/jmm.0.001817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 04/07/2024] Open
Abstract
There is growing evidence that altered microbiota abundance of a range of specific anaerobic bacteria are associated with cancer, including Peptoniphilus spp., Porphyromonas spp., Fusobacterium spp., Fenollaria spp., Prevotella spp., Sneathia spp., Veillonella spp. and Anaerococcus spp. linked to multiple cancer types. In this review we explore these pathogenic associations. The mechanisms by which bacteria are known or predicted to interact with human cells are reviewed and we present an overview of the interlinked mechanisms and hypotheses of how multiple intracellular anaerobic bacterial pathogens may act together to cause host cell and tissue microenvironment changes associated with carcinogenesis and cancer cell invasion. These include combined effects on changes in cell signalling, DNA damage, cellular metabolism and immune evasion. Strategies for early detection and eradication of anaerobic cancer-associated bacterial pathogens that may prevent cancer progression are proposed.
Collapse
Affiliation(s)
- Rachel Hurst
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Daniel S. Brewer
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- Earlham Institute, Norwich Research Park Innovation Centre, Colney Lane, Norwich NR4 7UZ, UK
| | - Abraham Gihawi
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - John Wain
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- Quadram Institute Biosciences, Colney Lane, Norwich, Norfolk, NR4 7UQ, UK
| | - Colin S. Cooper
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| |
Collapse
|
25
|
Chung IY, Kim J, Koh A. The Microbiome Matters: Its Impact on Cancer Development and Therapeutic Responses. J Microbiol 2024; 62:137-152. [PMID: 38587593 DOI: 10.1007/s12275-024-00110-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 04/09/2024]
Abstract
In the evolving landscape of cancer research, the human microbiome emerges as a pivotal determinant reshaping our understanding of tumorigenesis and therapeutic responses. Advanced sequencing technologies have uncovered a vibrant microbial community not confined to the gut but thriving within tumor tissues. Comprising bacteria, viruses, and fungi, this diverse microbiota displays distinct signatures across various cancers, with most research primarily focusing on bacteria. The correlations between specific microbial taxa within different cancer types underscore their pivotal roles in driving tumorigenesis and influencing therapeutic responses, particularly in chemotherapy and immunotherapy. This review amalgamates recent discoveries, emphasizing the translocation of the oral microbiome to the gut as a potential marker for microbiome dysbiosis across diverse cancer types and delves into potential mechanisms contributing to cancer promotion. Furthermore, it highlights the adverse effects of the microbiome on cancer development while exploring its potential in fortifying strategies for cancer prevention and treatment.
Collapse
Affiliation(s)
- In-Young Chung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| | - Jihyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
26
|
Feng P, Xue X, Bukhari I, Qiu C, Li Y, Zheng P, Mi Y. Gut microbiota and its therapeutic implications in tumor microenvironment interactions. Front Microbiol 2024; 15:1287077. [PMID: 38322318 PMCID: PMC10844568 DOI: 10.3389/fmicb.2024.1287077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
The development of cancer is not just the growth and proliferation of a single transformed cell, but its tumor microenvironment (TME) also coevolves with it, which is primarily involved in tumor initiation, development, metastasis, and therapeutic responses. Recent years, TME has been emerged as a potential target for cancer diagnosis and treatment. However, the clinical efficacy of treatments targeting the TME, especially its specific components, remains insufficient. In parallel, the gut microbiome is an essential TME component that is crucial in cancer immunotherapy. Thus, assessing and constructing frameworks between the gut microbiota and the TME can significantly enhance the exploration of effective treatment strategies for various tumors. In this review the role of the gut microbiota in human cancers, including its function and relationship with various tumors was summarized. In addition, the interaction between the gut microbiota and the TME as well as its potential applications in cancer therapeutics was described. Furthermore, it was summarized that fecal microbiota transplantation, dietary adjustments, and synthetic biology to introduce gut microbiota-based medical technologies for cancer treatment. This review provides a comprehensive summary for uncovering the mechanism underlying the effects of the gut microbiota on the TME and lays a foundation for the development of personalized medicine in further studies.
Collapse
Affiliation(s)
- Pengya Feng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Children Rehabilitation Medicine, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xia Xue
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ihtisham Bukhari
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunjing Qiu
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Li
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Peters BA, Kelly L, Wang T, Loudig O, Rohan TE. The Breast Microbiome in Breast Cancer Risk and Progression: A Narrative Review. Cancer Epidemiol Biomarkers Prev 2024; 33:9-19. [PMID: 37943168 DOI: 10.1158/1055-9965.epi-23-0965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/06/2023] [Accepted: 11/07/2023] [Indexed: 11/10/2023] Open
Abstract
A decade ago, studies in human populations first revealed the existence of a unique microbial community in the breast, a tissue historically viewed as sterile, with microbial origins seeded through the nipple and/or translocation from other body sites. Since then, research efforts have been made to characterize the microbiome in healthy and cancerous breast tissues. The purpose of this review is to summarize the current evidence for the association of the breast microbiome with breast cancer risk and progression. Briefly, while many studies have examined the breast microbiome in patients with breast cancer, and compared it with the microbiome of benign breast disease tissue or normal breast tissue, these studies have varied widely in their sample sizes, methods, and quality of evidence. Thus, while several large and rigorous cross-sectional studies have provided key evidence of an altered microbiome in breast tumors compared with normal adjacent and healthy control tissue, there are few consistent patterns of perturbed microbial taxa. In addition, only one large prospective study has provided evidence of a relationship between the breast tumor microbiota and cancer prognosis. Future research studies featuring large, well-characterized cohorts with prospective follow-up for breast cancer incidence, progression, and response to treatment are warranted.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Libusha Kelly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
28
|
Jiang S, Ma W, Ma C, Zhang Z, Zhang W, Zhang J. An emerging strategy: probiotics enhance the effectiveness of tumor immunotherapy via mediating the gut microbiome. Gut Microbes 2024; 16:2341717. [PMID: 38717360 PMCID: PMC11085971 DOI: 10.1080/19490976.2024.2341717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
The occurrence and progression of tumors are often accompanied by disruptions in the gut microbiota. Inversely, the impact of the gut microbiota on the initiation and progression of cancer is becoming increasingly evident, influencing the tumor microenvironment (TME) for both local and distant tumors. Moreover, it is even suggested to play a significant role in the process of tumor immunotherapy, contributing to high specificity in therapeutic outcomes and long-term effectiveness across various cancer types. Probiotics, with their generally positive influence on the gut microbiota, may serve as effective agents in synergizing cancer immunotherapy. They play a crucial role in activating the immune system to inhibit tumor growth. In summary, this comprehensive review aims to provide valuable insights into the dynamic interactions between probiotics, gut microbiota, and cancer. Furthermore, we highlight recent advances and mechanisms in using probiotics to improve the effectiveness of cancer immunotherapy. By understanding these complex relationships, we may unlock innovative approaches for cancer diagnosis and treatment while optimizing the effects of immunotherapy.
Collapse
Affiliation(s)
- Shuaiming Jiang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wenyao Ma
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Chenchen Ma
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, Shenzhen, PR China
| | - Zeng Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wanli Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| |
Collapse
|
29
|
Alvarez-Frutos L, Barriuso D, Duran M, Infante M, Kroemer G, Palacios-Ramirez R, Senovilla L. Multiomics insights on the onset, progression, and metastatic evolution of breast cancer. Front Oncol 2023; 13:1292046. [PMID: 38169859 PMCID: PMC10758476 DOI: 10.3389/fonc.2023.1292046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer is the most common malignant neoplasm in women. Despite progress to date, 700,000 women worldwide died of this disease in 2020. Apparently, the prognostic markers currently used in the clinic are not sufficient to determine the most appropriate treatment. For this reason, great efforts have been made in recent years to identify new molecular biomarkers that will allow more precise and personalized therapeutic decisions in both primary and recurrent breast cancers. These molecular biomarkers include genetic and post-transcriptional alterations, changes in protein expression, as well as metabolic, immunological or microbial changes identified by multiple omics technologies (e.g., genomics, epigenomics, transcriptomics, proteomics, glycomics, metabolomics, lipidomics, immunomics and microbiomics). This review summarizes studies based on omics analysis that have identified new biomarkers for diagnosis, patient stratification, differentiation between stages of tumor development (initiation, progression, and metastasis/recurrence), and their relevance for treatment selection. Furthermore, this review highlights the importance of clinical trials based on multiomics studies and the need to advance in this direction in order to establish personalized therapies and prolong disease-free survival of these patients in the future.
Collapse
Affiliation(s)
- Lucia Alvarez-Frutos
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Daniel Barriuso
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mercedes Duran
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mar Infante
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, Paris, France
| | - Roberto Palacios-Ramirez
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Laura Senovilla
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
30
|
Actis S, Cazzaniga M, Bounous VE, D'Alonzo M, Rosso R, Accomasso F, Minella C, Biglia N. Emerging evidence on the role of breast microbiota on the development of breast cancer in high-risk patients. Carcinogenesis 2023; 44:718-725. [PMID: 37793149 DOI: 10.1093/carcin/bgad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 08/02/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023] Open
Abstract
Cancer is a multi-factorial disease, and the etiology of breast cancer (BC) is due to a combination of both genetic and environmental factors. Breast tissue shows a unique microbiota, Proteobacteria and Firmicutes are the most abundant bacteria in breast tissue, and several studies have shown that the microbiota of healthy breast differs from that of BC. Breast microbiota appears to be correlated with different characteristics of the tumor, and prognostic clinicopathologic features. It also appears that there are subtle differences between the microbial profiles of the healthy control and high-risk patients. Genetic predisposition is an extremely important risk factor for BC. BRCA1/2 germline mutations and Li-Fraumeni syndrome are DNA repair deficiency syndromes inherited as autosomal dominant characters that substantially increase the risk of BC. These syndromes exhibit incomplete penetrance of BC expression in carrier subjects. The action of breast microbiota on carcinogenesis might explain why women with a mutation develop cancer and others do not. Among the potential biological pathways through which the breast microbiota may affect tumorigenesis, the most relevant appear to be DNA damage caused by colibactin and other bacterial-derived genotoxins, β-glucuronidase-mediated estrogen deconjugation and reactivation, and HPV-mediated cancer susceptibility. In conclusion, in patients with a genetic predisposition, an unfavorable breast microbiota may be co-responsible for the onset of BC. Prospectively, the ability to modulate the microbiota may have an impact on disease onset and progression in patients at high risk for BC.
Collapse
Affiliation(s)
- Silvia Actis
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| | | | - Valentina Elisabetta Bounous
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| | - Marta D'Alonzo
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| | - Roberta Rosso
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| | - Francesca Accomasso
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| | - Carola Minella
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| | - Nicoletta Biglia
- Gynecology and Obstetrics Unit, Department of Surgical Sciences, Mauriziano Umberto I Hospital, University of Turin, 10128 Turin, Italy
| |
Collapse
|
31
|
Kaźmierczak-Siedlecka K, Bulman N, Ulasiński P, Sobocki BK, Połom K, Marano L, Kalinowski L, Skonieczna-Żydecka K. Pharmacomicrobiomics of cell-cycle specific anti-cancer drugs - is it a new perspective for personalized treatment of cancer patients? Gut Microbes 2023; 15:2281017. [PMID: 37985748 PMCID: PMC10730203 DOI: 10.1080/19490976.2023.2281017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
Intestinal bacteria are equipped with an enzyme apparatus that is involved in the active biotransformation of xenobiotics, including drugs. Pharmacomicrobiomics, a new area of pharmacology, analyses interactions between bacteria and xenobiotics. However, there is another side to the coin. Pharmacotherapeutic agents can significantly modify the microbiota, which consequently affects their efficacy. In this review, we comprehensively gathered scientific evidence on the interplay between anticancer therapies and gut microbes. We also underlined how such interactions might impact the host response to a given therapy. We discuss the possibility of modulating the gut microbiota to increase the effectiveness/decrease the incidence of adverse events during tumor therapy. The anticipation of the future brings new evidence that gut microbiota is a target of interest to increase the efficacy of therapy.
Collapse
Affiliation(s)
- Karolina Kaźmierczak-Siedlecka
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
| | - Nikola Bulman
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
| | - Paweł Ulasiński
- Unit of Surgery with Unit of Oncological Surgery in Koscierzyna, Kościerzyna, Poland
| | - Bartosz Kamil Sobocki
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdańsk, Poland
| | - Karol Połom
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
| | - Luigi Marano
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | | |
Collapse
|
32
|
Ruiz-Saavedra S, Zapico A, González S, Salazar N, de los Reyes-Gavilán CG. Role of the intestinal microbiota and diet in the onset and progression of colorectal and breast cancers and the interconnection between both types of tumours. MICROBIOME RESEARCH REPORTS 2023; 3:6. [PMID: 38455079 PMCID: PMC10917624 DOI: 10.20517/mrr.2023.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/12/2023] [Accepted: 11/21/2023] [Indexed: 03/09/2024]
Abstract
Colorectal cancer (CRC) is among the leading causes of mortality in adults of both sexes worldwide, while breast cancer (BC) is among the leading causes of death in women. In addition to age, gender, and genetic predisposition, environmental and lifestyle factors exert a strong influence. Global diet, including alcohol consumption, is one of the most important modifiable factors affecting the risk of CRC and BC. Western dietary patterns promoting high intakes of xenobiotics from food processing and ethanol have been associated with increased cancer risk, whereas the Mediterranean diet, generally leading to a higher intake of polyphenols and fibre, has been associated with a protective effect. Gut dysbiosis is a common feature in CRC, where the usual microbiota is progressively replaced by opportunistic pathogens and the gut metabolome is altered. The relationship between microbiota and BC has been less studied. The estrobolome is the collection of genes from intestinal bacteria that can metabolize oestrogens. In a dysbiosis condition, microbial deconjugating enzymes can reactivate conjugated-deactivated oestrogens, increasing the risk of BC. In contrast, intestinal microorganisms can increase the biological activity and bioavailability of dietary phytochemicals through diverse microbial metabolic transformations, potentiating their anticancer activity. Members of the intestinal microbiota can increase the toxicity of xenobiotics through metabolic transformations. However, most of the microorganisms involved in diet-microbiota interactions remain poorly characterized. Here, we provide an overview of the associations between microbiota and diet in BC and CRC, considering the diverse types and heterogeneity of these cancers and their relationship between them and with gut microbiota.
Collapse
Affiliation(s)
- Sergio Ruiz-Saavedra
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa 33300, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Aida Zapico
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
- Department of Functional Biology, University of Oviedo, Oviedo 33006, Spain
| | - Sonia González
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
- Department of Functional Biology, University of Oviedo, Oviedo 33006, Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa 33300, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa 33300, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| |
Collapse
|
33
|
Khatri VA, Paul S, Patel NJ, Thippani S, Sawant JY, Durkee KL, Murphy CL, Aleman GO, Valentino JA, Jathan J, Melillo A, Sapi E. Global transcriptomic analysis of breast cancer and normal mammary epithelial cells infected with Borrelia burgdorferi. Eur J Microbiol Immunol (Bp) 2023; 13:63-76. [PMID: 37856211 PMCID: PMC10668924 DOI: 10.1556/1886.2023.00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
The bacterial spirochete Borrelia burgdorferi, the causative agent of Lyme Disease, can disseminate and colonize various tissues and organs, orchestrating severe clinical symptoms including arthritis, carditis, and neuroborreliosis. Previous research has demonstrated that breast cancer tissues could provide an ideal habitat for diverse populations of bacteria, including B. burgdorferi, which is associated with a poor prognosis. Recently, we demonstrated that infection with B. burgdorferi enhances the invasion and migration of triple-negative MDA-MB-231 cells which represent a type of breast tumor with more aggressive cancer traits. In this study, we hypothesized that infection by B. burgdorferi affects the expression of cancer-associated genes to effectuate breast cancer phenotypes. We applied the high-throughput technique of RNA-sequencing on B. burgdorferi-infected MDA-MB-231 breast cancer and normal-like MCF10A cells to determine the most differentially expressed genes (DEG) upon infection. Overall, 142 DEGs were identified between uninfected and infected samples in MDA-MB-231 while 95 DEGs were found in MCF10A cells. A major trend of the upregulation of C-X-C and C-C motif chemokine family members as well as genes and pathways was associated with infection, inflammation, and cancer. These genes could serve as potential biomarkers for pathogen-related tumorigenesis and cancer progression which could lead to new therapeutic opportunities.
Collapse
Affiliation(s)
- Vishwa A. Khatri
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Sambuddha Paul
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Niraj Jatin Patel
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Sahaja Thippani
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Janhavi Y. Sawant
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Katie L. Durkee
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Cassandra L. Murphy
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Geneve Ortiz Aleman
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Justine A. Valentino
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Jasmine Jathan
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Anthony Melillo
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| | - Eva Sapi
- Lyme Disease Research Group, Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Road, West Haven, CT 06516, USA
| |
Collapse
|
34
|
Jotshi A, Sukla KK, Haque MM, Bose C, Varma B, Koppiker CB, Joshi S, Mishra R. Exploring the human microbiome - A step forward for precision medicine in breast cancer. Cancer Rep (Hoboken) 2023; 6:e1877. [PMID: 37539732 PMCID: PMC10644338 DOI: 10.1002/cnr2.1877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/24/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The second most frequent cancer in the world and the most common malignancy in women is breast cancer. Breast cancer is a significant health concern in India with a high mortality-to-incidence ratio and presentation at a younger age. RECENT FINDINGS Recent studies have identified gut microbiota as a significant factor that can have an influence on the development, treatment, and prognosis of breast cancer. This review article aims to describe the influence of microbial dysbiosis on breast cancer occurrence and the possible interactions between oncobiome and specific breast cancer molecular subtypes. The review further also discusses the role of epigenetics and diet/nutrition in the regulation of the gut and breast microbiome and its association with breast cancer prevention, therapy, and recurrence. Additionally, the recent technological advances in microbiome research, including next-generation sequencing (NGS) technologies, genome sequencing, single-cell sequencing, and microbial metabolomics along with recent advances in artificial intelligence (AI) have also been reviewed. This is an attempt to present a comprehensive status of the microbiome as a key cancer biomarker. CONCLUSION We believe that correlating microbiome and carcinogenesis is important as it can provide insights into the mechanisms by which microbial dysbiosis can influence cancer development and progression, leading to the potential use of the microbiome as a tool for prognostication and personalized therapy.
Collapse
Affiliation(s)
- Asmita Jotshi
- Centre for Translational Cancer Research: A Joint Initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM)PuneIndia
| | | | | | - Chandrani Bose
- Life Sciences R&D, TCS Research, Tata Consultancy Services LimitedPuneIndia
| | - Binuja Varma
- TCS Genomics Lab, Tata Consultancy Services LimitedNew DelhiIndia
| | - C. B. Koppiker
- Centre for Translational Cancer Research: A Joint Initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM)PuneIndia
- Prashanti Cancer Care Mission, Pune, India and Orchids Breast Health Centre, a PCCM initiativePuneIndia
| | - Sneha Joshi
- Centre for Translational Cancer Research: A Joint Initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM)PuneIndia
| | - Rupa Mishra
- Centre for Translational Cancer Research: A Joint Initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM)PuneIndia
| |
Collapse
|
35
|
Singla RK, Wang X, Gundamaraju R, Joon S, Tsagkaris C, Behzad S, Khan J, Gautam R, Goyal R, Rakmai J, Dubey AK, Simal-Gandara J, Shen B. Natural products derived from medicinal plants and microbes might act as a game-changer in breast cancer: a comprehensive review of preclinical and clinical studies. Crit Rev Food Sci Nutr 2023; 63:11880-11924. [PMID: 35838143 DOI: 10.1080/10408398.2022.2097196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Breast cancer (BC) is the most prevalent neoplasm among women. Genetic and environmental factors lead to BC development and on this basis, several preventive - screening and therapeutic interventions have been developed. Hormones, both in the form of endogenous hormonal signaling or hormonal contraceptives, play an important role in BC pathogenesis and progression. On top of these, breast microbiota includes both species with an immunomodulatory activity enhancing the host's response against cancer cells and species producing proinflammatory cytokines associated with BC development. Identification of novel multitargeted therapeutic agents with poly-pharmacological potential is a dire need to combat advanced and metastatic BC. A growing body of research has emphasized the potential of natural compounds derived from medicinal plants and microbial species as complementary BC treatment regimens, including dietary supplements and probiotics. In particular, extracts from plants such as Artemisia monosperma Delile, Origanum dayi Post, Urtica membranacea Poir. ex Savigny, Krameria lappacea (Dombey) Burdet & B.B. Simpson and metabolites extracted from microbes such as Deinococcus radiodurans and Streptomycetes strains as well as probiotics like Bacillus coagulans and Lactobacillus brevis MK05 have exhibited antitumor effects in the form of antiproliferative and cytotoxic activity, increase in tumors' chemosensitivity, antioxidant activity and modulation of BC - associated molecular pathways. Further, bioactive compounds like 3,3'-diindolylmethane, epigallocatechin gallate, genistein, rutin, resveratrol, lycopene, sulforaphane, silibinin, rosmarinic acid, and shikonin are of special interest for the researchers and clinicians because these natural agents have multimodal action and act via multiple ways in managing the BC and most of these agents are regularly available in our food and fruit diets. Evidence from clinical trials suggests that such products had major potential in enhancing the effectiveness of conventional antitumor agents and decreasing their side effects. We here provide a comprehensive review of the therapeutic effects and mechanistic underpinnings of medicinal plants and microbial metabolites in BC management. The future perspectives on the translation of these findings to the personalized treatment of BC are provided and discussed.
Collapse
Affiliation(s)
- Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Xiaoyan Wang
- Department of Pathology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Lab, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Shikha Joon
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | | | - Sahar Behzad
- Evidence-based Phytotherapy and Complementary Medicine Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | - Rupesh Gautam
- Department of Pharmacology, MM School of Pharmacy, MM University, Sadopur, Haryana, India
| | - Rajat Goyal
- Department of Pharmacology, MM School of Pharmacy, MM University, Sadopur, Haryana, India
| | - Jaruporn Rakmai
- Kasetsart Agricultural and Agro-Industrial Product Improvement Institute (KAPI), Kasetsart University, Bangkok, Thailand
| | | | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade de Vigo, Ourense, Spain
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Chapadgaonkar SS, Bajpai SS, Godbole MS. Gut microbiome influences incidence and outcomes of breast cancer by regulating levels and activity of steroid hormones in women. Cancer Rep (Hoboken) 2023; 6:e1847. [PMID: 37311575 PMCID: PMC10644331 DOI: 10.1002/cnr2.1847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/21/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Breast cancer, the leading cancer type in women worldwide, is affected by reproductive and nonreproductive factors. Estrogen and progesterone influence the incidence and progression of breast cancer. The microbiome of the gut, a complex organ that plays a vital role in digestion and homeostasis, enhances availability of estrogen and progesterone in the host. Thus, an altered gut microbiome may influence the hormone-induced breast cancer incidence. This review describes the current understanding of the roles of gut microbiome in influencing the incidence and progression of breast cancer, with an emphasis on the microbiome-induced metabolism of estrogen and progesterone. RECENT FINDINGS Microbiome has been recognized as a promising hallmark of cancer. Next-generation sequencing technologies have aided in rapid identification of components of the gut microbiome that are capable of metabolizing estrogen and progesterone. Moreover, studies have indicated a wider role of the gut microbiome in metabolizing chemotherapeutic and hormonal therapy agents and reducing their efficacy in patients with breast cancer, with a predominant effect in postmenopausal women. CONCLUSION The gut microbiome and variations in its composition significantly alter the incidence and therapy outcomes of patients with breast cancer. Thus, a healthy and diverse microbiome is required for better response to anticancer therapies. Finally, the review emphasizes the requirement of studies to elucidate mechanisms that may aid in improving the gut microbiome composition, and hence, survival outcomes of patients with breast cancer.
Collapse
Affiliation(s)
- Shilpa S. Chapadgaonkar
- Department of Biosciences and Technology, Faculty of Sciences and Health SciencesDr. Vishwanath Karad MIT World Peace UniversityPuneIndia
| | - Srashti S. Bajpai
- Department of Biosciences and Technology, Faculty of Sciences and Health SciencesDr. Vishwanath Karad MIT World Peace UniversityPuneIndia
| | - Mukul S. Godbole
- Department of Biosciences and Technology, Faculty of Sciences and Health SciencesDr. Vishwanath Karad MIT World Peace UniversityPuneIndia
| |
Collapse
|
37
|
Gao X, Yang H, Chu Y, Zhang W, Wang Z, Ji L. The specific viral composition in triple-negative breast cancer tissue shapes the specific tumor microenvironment characterized on pathological images. Microb Pathog 2023; 184:106385. [PMID: 37813319 DOI: 10.1016/j.micpath.2023.106385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
Numerous studies have shown that different subtypes of breast cancer (BC) have significant differences in terms of the tumor microbiome, host gene expression, and histopathological image, whereas the biological links between these cancer-associated indicators are still unknown. Here, we performed a comprehensive analysis with 610 patients of the four subtypes of BC with matched tissue microbiota, host transcriptome, and histopathological image samples. Correlation analysis showed that the composition of intratumoral viruses shaped the tumor microenvironment (TME) of patients with BC, and the TME was further reflected in the histopathological images. Of the four subtypes, patients with triple-negative breast cancer (TNBC) had unique intratumoral viral community composition, non-cancer cell infiltration in the TME, and histopathological image characteristics. Furthermore, we detected multiple virus-cell-image association axes in TNBC, in which tumor-associated macrophages (TAMs) have clinical prognostic implication. This study provides a comprehensive map of the associations between the intratumoral virome, TME, and histopathological image of TNBC, as well as insights into disease prognosis that can be crucial for precise therapeutic intervention strategies.
Collapse
Affiliation(s)
- Xuzhu Gao
- Institute of Clinical Oncology, The Second People's Hospital of Lianyungang City (Cancer Hospital of Lianyungang), Lianyungang, China; Department of Central Laboratory, Lianyungang Hospital Affiliated to Kangda College of Nanjing Medical University, Lianyungang, China
| | - Hailong Yang
- Geneis Beijing Co., Ltd., Beijing, 100102, China; Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, 266000, China; School of Electrical & Information Engineering, Anhui University of Technology, Anhui, 243002, China
| | - Yuwen Chu
- Geneis Beijing Co., Ltd., Beijing, 100102, China; Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, 266000, China; School of Electrical & Information Engineering, Anhui University of Technology, Anhui, 243002, China
| | - Wenjing Zhang
- Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Zhongchen Wang
- Department of General Surgery, Daqing Longnan Hospital, The Fifth Affiliated Hospital of Qiqihar Medical College, Daqing, 163453, China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing, 100102, China; Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, 266000, China.
| |
Collapse
|
38
|
Liu K, Jia N, Shi H, Ran Y. Current and future research on the association between gut microbiota and breast cancer. Front Microbiol 2023; 14:1272275. [PMID: 38029117 PMCID: PMC10646191 DOI: 10.3389/fmicb.2023.1272275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Breast cancer (BC) is a prevalent malignancy. There exists a strong association between gut microbiota (GM) and the development of BC. The GM composition in individuals with BC significantly differs from that in their healthy counterparts. Furthermore, the distribution of GM varies significantly among individuals with different types of BC. The GM can impact BC through metabolite secretion, the gut-mammary axis, and other pathways. Modulating the GM can serve as a very promising potential therapeutic strategy in the treatment of BC. This article will summarize existing research, focusing on the relationship between intestinal microbiota and BC. At the same time, the project will also analyze the application value of intestinal microorganisms in BC intervention work, so as to provide a reference for the further development of BC prevention and treatment work.
Collapse
Affiliation(s)
| | - Nan Jia
- Affiliated Hospital of Hebei University, Baoding, China
| | - Hongyun Shi
- Affiliated Hospital of Hebei University, Baoding, China
| | - Yuge Ran
- Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
39
|
Wang Y, Qu D, Zhang Y, Jin Y, Feng Y, Zhang H, Xia Q. Intra-tumoral microbial community profiling and associated metabolites alterations of TNBC. Front Oncol 2023; 13:1143163. [PMID: 37901331 PMCID: PMC10602718 DOI: 10.3389/fonc.2023.1143163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Triple-negative breast cancer (TNBC) presents significant challenges to female health owing to the lack of therapeutic targets and its poor prognosis. In recent years, in the field of molecular pathology, there has been a growing focus on the role of intra-tumoral microbial communities and metabolic alterations in tumor cells. However, the precise mechanism through which microbiota and their metabolites influence TNBC remains unclear and warrants further investigation. In this study, we analyzed the microbial community composition in various subtypes of breast cancer through 16S rRNA MiSeq sequencing of formalin-fixed, paraffin-embedded (FFPE) tissue samples. Notably, Turicibacter, a microbe associated with cancer response, exhibited a significantly higher abundance in TNBC. Similarly, mass spectrometry-based metabolomic analysis revealed substantial differences in specific metabolites, such as nutriacholic, pregnanetriol, and cortol. Furthermore, we observed significant correlations between the intra-tumoral microbiome, clinicopathological characteristics, and human epidermal growth factor receptor-2 expression(HER2). Three microbial taxa (Cytophagaceae, Conexibacteraceae, and Flavobacteriaceae) were associated with tumor-infiltrating lymphocytes(TILs), which are indicative of antitumor immunity. This study creatively utilized FFPE tissue samples to assess intra-tumoral microbial communities and their related metabolic correlations, presenting avenues for the identification of novel diagnostic biomarkers, the development of therapeutic strategies, and the early clinical diagnosis of TNBC.
Collapse
Affiliation(s)
- Yi Wang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, China
- Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, China
| | - Dingding Qu
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, China
- Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, China
| | - Yali Zhang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, China
- Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, China
| | - Yiping Jin
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, China
- Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, China
| | - Yu Feng
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - He Zhang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, China
- Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, China
| | - Qingxin Xia
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, China
- Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, China
| |
Collapse
|
40
|
Desalegn Z, Smith A, Yohannes M, Cao X, Anberber E, Bekuretsion Y, Assefa M, Bauer M, Vetter M, Kantelhardt EJ, Abebe T, Starlard-Davenport A. Human Breast Tissue Microbiota Reveals Unique Microbial Signatures that Correlate with Prognostic Features in Adult Ethiopian Women with Breast Cancer. Cancers (Basel) 2023; 15:4893. [PMID: 37835588 PMCID: PMC10571711 DOI: 10.3390/cancers15194893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) is the leading cause of cancer mortality among women in Ethiopia. Overall, women of African ancestry have the highest death toll due to BC compared to other racial/ethnic groups. The cause of the disparity in mortality is unclear. Recently, studies conducted in the United States and other high-income countries highlighted the role of microbial dysbiosis in BC initiation, tumor growth, and treatment outcome. However, the extent to which inter-individual differences in the makeup of microbiota are associated with clinical and histopathological outcomes in Ethiopian women has not been studied. The goal of our study was to profile the microbiome in breast tumor and normal adjacent to tumor (NAT) tissues of the same donor and to identify associations between microbial composition and abundance and clinicopathological factors in Ethiopian women with BC. We identified 14 microbiota genera in breast tumor tissues that were distinct from NAT tissues, of which Sphingobium, Anaerococcus, Corynebacterium, Delftia, and Enhydrobacter were most significantly decreased in breast tumors compared to NAT tissues. Several microbial genera significantly differed by clinicopathological factors in Ethiopian women with BC. Specifically, the genus Burkholderia more strongly correlated with aggressive triple negative (TNBC) and basal-like breast tumors. The genera Alkanindiges, Anoxybacillus, Leifsonia, and Exiguobacterium most strongly correlated with HER2-E tumors. Luminal A and luminal B tumors also correlated with Anoxybacillus but not as strongly as HER2-E tumors. A relatively higher abundance of the genus Citrobacter most significantly correlated with advanced-stage breast tumors compared to early-stage tumors. This is the first study to report an association between breast microbial dysbiosis and clinicopathological factors in Ethiopian women.
Collapse
Affiliation(s)
- Zelalem Desalegn
- Department of Microbiology, Immunology, and Parasitology, School of Medicine, College of Health Sciences Addis Ababa University, Addis Ababa 9086, Ethiopia; (Z.D.); (M.Y.); (T.A.)
- Global Health Working Group, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
| | - Alana Smith
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Meron Yohannes
- Department of Microbiology, Immunology, and Parasitology, School of Medicine, College of Health Sciences Addis Ababa University, Addis Ababa 9086, Ethiopia; (Z.D.); (M.Y.); (T.A.)
- Global Health Working Group, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
- School of Medical Laboratory Sciences, Addis Ababa University, Addis Ababa 9086, Ethiopia;
| | - Xueyuan Cao
- Department of Health Promotion and Disease Prevention, College of Nursing, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Endale Anberber
- Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa 9086, Ethiopia;
| | - Yonas Bekuretsion
- Department of Pathology, School of Medicine, Addis Ababa University, Addis Ababa 9086, Ethiopia;
| | - Mathewos Assefa
- Department of Oncology, School of Medicine, Addis Ababa University, Addis Ababa 9086, Ethiopia;
| | - Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
| | - Martina Vetter
- Department of Gynecology, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
| | - Eva Johanna Kantelhardt
- Global Health Working Group, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
- Department of Gynecology, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany
| | - Tamrat Abebe
- Department of Microbiology, Immunology, and Parasitology, School of Medicine, College of Health Sciences Addis Ababa University, Addis Ababa 9086, Ethiopia; (Z.D.); (M.Y.); (T.A.)
- Global Health Working Group, Martin Luther University Halle-Wittenberg, 06097 Halle (Saale), Germany;
| | - Athena Starlard-Davenport
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| |
Collapse
|
41
|
Alwehaibi MA, Al-Ansari MM, Alfadda AA, Al-Malki R, Masood A, Abdel Rahman AM, Benabdelkamel H. Proteomics Investigation of the Impact of the Enterococcus faecalis Secretome on MCF-7 Tumor Cells. Int J Mol Sci 2023; 24:14937. [PMID: 37834385 PMCID: PMC10573200 DOI: 10.3390/ijms241914937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer is the most prevalent form of cancer among women. The microenvironment of a cancer tumor is surrounded by various cells, including the microbiota. An imbalance between microbes and their host may contribute to the development and spread of breast cancer. Therefore, the objective of this study is to investigate the influence of Enterococcus faecalis on a breast cancer cell line (MCF-7) to mimic the luminal A subtype of breast cancer, using an untargeted proteomics approach to analyze the proteomic profiles of breast cancer cells after their treatment with E. faecalis in order to understand the microbiome and its role in the development of cancer. The breast cancer cell line MCF-7 was cultured and then treated with a 10% bacterial supernatant at two time points (24 h and 48 h) at 37 °C in a humidified incubator with 5% CO2. Proteins were then extracted and separated using two-dimensional difference (2D-DIGE) gel electrophoresis, and the statistically significant proteins (p-value < 0.05, fold change > 1.5) were identified via matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF-MS). The protein fingerprints showed a differential protein expression pattern in the cells treated with E. faecalis for 24 and 48 h compared with the control. We found 58 statistically significant proteins changes in the MCF-7 breast cancer cells affected by E. faecalis. Kilin and transgelin were upregulated after 24 h of treatment and could be used as diagnostic and prognostic markers for breast cancer. In addition, another protein involved in the inhibition of cell proliferation was coiled-coil domain-containing protein 154. The protein markers identified in this study may serve as possible biomarkers for breast cancer progression. This promotes their future uses as important therapeutic goals in the treatment and diagnosis of cancer and increases our understanding of the breast microbiome and its role in the development of cancer.
Collapse
Affiliation(s)
- Moudi A Alwehaibi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Mysoon M Al-Ansari
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Assim A Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, Riyadh 11451, Saudi Arabia
| | - Reem Al-Malki
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Anas M Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| |
Collapse
|
42
|
Wu Y, Zhang Y, Zhang W, Huang Y, Lu X, Shang L, Zhou Z, Chen X, Li S, Cheng S, Song Y. The tremendous clinical potential of the microbiota in the treatment of breast cancer: the next frontier. J Cancer Res Clin Oncol 2023; 149:12513-12534. [PMID: 37382675 DOI: 10.1007/s00432-023-05014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Although significant advances have been made in the diagnosis and treatment of breast cancer (BC) in recent years, BC remains the most common cancer in women and one of the main causes of death among women worldwide. Currently, more than half of BC patients have no known risk factors, emphasizing the significance of identifying more tumor-related factors. Therefore, we urgently need to find new therapeutic strategies to improve prognosis. Increasing evidence demonstrates that the microbiota is present in a wider range of cancers beyond colorectal cancer. BC and breast tissues also have different types of microbiotas that play a key role in carcinogenesis and in modulating the efficacy of anticancer treatment, for instance, chemotherapy, radiotherapy, and immunotherapy. In recent years, studies have confirmed that the microbiota can be an important factor directly and/or indirectly affecting the occurrence, metastasis and treatment of BC by regulating different biological processes, such as estrogen metabolism, DNA damage, and bacterial metabolite production. Here, we review the different microbiota-focused studies associated with BC and explore the mechanisms of action of the microbiota in BC initiation and metastasis and its application in various therapeutic strategies. We found that the microbiota has vital clinical value in the diagnosis and treatment of BC and could be used as a biomarker for prognosis prediction. Therefore, modulation of the gut microbiota and its metabolites might be a potential target for prevention or therapy in BC.
Collapse
Affiliation(s)
- Yang Wu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Yue Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenwen Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuanxi Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Xiangshi Lu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Lingmin Shang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Zhaoyue Zhou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Xiaolu Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Shuhui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Shaoqiang Cheng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China.
| | - Yanni Song
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
43
|
Algrafi AS, Jamal AA, Ismaeel DM. Microbiota as a New Target in Cancer Pathogenesis and Treatment. Cureus 2023; 15:e47072. [PMID: 38021696 PMCID: PMC10645418 DOI: 10.7759/cureus.47072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 12/01/2023] Open
Abstract
The microbial ecosystem of humans is an integral part of human health and disease. A significant percentage of tumors worldwide are thought to be microbially induced. The relationship between cancer and microbes is complex. In this article review, we aim to give an overview of human microbiota and its role in carcinogenesis, emphasize the relation between microbiota and cancer immunity, and highlight its role in the future of cancer therapy. The term microbiota refers to the collection of microorganisms that are located in an individual, whereas the total genome of these microorganisms is referred to as the microbiome. The microbiota in humans has many physiological functions. The microbiota within the gut lumen has a profound effect on the local and systemic immune system. The immune system can change the gut microbiota. Microbiota may induce carcinogenesis by several mechanisms. It also affects tumor progression. Thus, microbiota modulation may aid in the prevention and treatment of cancer. Intentionally introducing microorganisms into the oncological patient is assumed to mobilize the immune system to become able to, at least, limit the development of cancer. Microbes are used as vectors which are carriers of particular antineoplastic agents that reduce the side effects of chemotherapy. Inflammation and tumor microenvironment play an essential role in promoting chemo-resistance. There is now considerable evidence, both in humans as well as in laboratory animals, that the commensal microbiota has important effects on carcinogenesis, tumor growth, and therapy response.
Collapse
Affiliation(s)
- Abeer S Algrafi
- Internal Medicine, College of Medicine, Taibah University, Madinah, SAU
| | - Aisha A Jamal
- General Practice, College of Medicine, Taibah University, Madinah, SAU
| | - Dana M Ismaeel
- General Practice, College of Medicine, Taibah University, Madinah, SAU
| |
Collapse
|
44
|
Li D, Zhu L, Wang Y, Zhou X, Li Y. Bacterial outer membrane vesicles in cancer: Biogenesis, pathogenesis, and clinical application. Biomed Pharmacother 2023; 165:115120. [PMID: 37442066 DOI: 10.1016/j.biopha.2023.115120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/18/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
Outer membrane vesicles (OMVs) are spherical, nano-sized particles of bilayer lipid structure secreted by Gram-negative bacteria. They contain a series of cargos from bacteria and are important messengers for communication between bacteria and their environment. OMVs play multiple roles in bacterial survival and adaptation and can affect host physiological functions and disease development by acting on host cell membranes and altering host cell signaling pathways. This paper summarizes the mechanisms of OMV genesis and the multiple roles of OMVs in the tumor microenvironment. Also, this paper discusses the prospects of OMVs for a wide range of applications in drug delivery, tumor diagnosis, and therapy.
Collapse
Affiliation(s)
- Deming Li
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China
| | - Lisi Zhu
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China
| | - Yuxiao Wang
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China
| | - Xiangyu Zhou
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China.
| | - Yan Li
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China.
| |
Collapse
|
45
|
Xue X, Li R, Chen Z, Li G, Liu B, Guo S, Yue Q, Yang S, Xie L, Zhang Y, Zhao J, Tan R. The role of the symbiotic microecosystem in cancer: gut microbiota, metabolome, and host immunome. Front Immunol 2023; 14:1235827. [PMID: 37691931 PMCID: PMC10484231 DOI: 10.3389/fimmu.2023.1235827] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/12/2023] [Indexed: 09/12/2023] Open
Abstract
The gut microbiota is not just a simple nutritional symbiosis that parasitizes the host; it is a complex and dynamic ecosystem that coevolves actively with the host and is involved in a variety of biological activities such as circadian rhythm regulation, energy metabolism, and immune response. The development of the immune system and immunological functions are significantly influenced by the interaction between the host and the microbiota. The interactions between gut microbiota and cancer are of a complex nature. The critical role that the gut microbiota plays in tumor occurrence, progression, and treatment is not clear despite the already done research. The development of precision medicine and cancer immunotherapy further emphasizes the importance and significance of the question of how the microbiota takes part in cancer development, progression, and treatment. This review summarizes recent literature on the relationship between the gut microbiome and cancer immunology. The findings suggest the existence of a "symbiotic microecosystem" formed by gut microbiota, metabolome, and host immunome that is fundamental for the pathogenesis analysis and the development of therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Xiaoyu Xue
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenni Chen
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
- College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Guiyu Li
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Bisheng Liu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shanshan Guo
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Qianhua Yue
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Siye Yang
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Linlin Xie
- Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Classical Chinese Medicine Diagnosis and Treatment Center, Luzhou, China
| | - Yiguan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Junning Zhao
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Ruirong Tan
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
46
|
Hoskinson C, Jiang RY, Stiemsma LT. Elucidating the roles of the mammary and gut microbiomes in breast cancer development. Front Oncol 2023; 13:1198259. [PMID: 37664075 PMCID: PMC10470065 DOI: 10.3389/fonc.2023.1198259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
The mammary microbiome is a newly characterized bacterial niche that might offer biological insight into the development of breast cancer. Together with in-depth analysis of the gut microbiome in breast cancer, current evidence using next-generation sequencing and metabolic profiling suggests compositional and functional shifts in microbial consortia are associated with breast cancer. In this review, we discuss the fundamental studies that have progressed this important area of research, focusing on the roles of both the mammary tissue microbiome and the gut microbiome. From the literature, we identified the following major conclusions, (I) There are unique breast and gut microbial signatures (both compositional and functional) that are associated with breast cancer, (II) breast and gut microbiome compositional and breast functional dysbiosis represent potential early events of breast tumor development, (III) specific breast and gut microbes confer host immune responses that can combat breast tumor development and progression, and (IV) chemotherapies alter the microbiome and thus maintenance of a eubiotic microbiome may be key in breast cancer treatment. As the field expectantly advances, it is necessary for the role of the microbiome to continue to be elucidated using multi-omic approaches and translational animal models in order to improve predictive, preventive, and therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Courtney Hoskinson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | | | - Leah T. Stiemsma
- Natural Science Division, Pepperdine University, Malibu, CA, United States
| |
Collapse
|
47
|
AlMalki RH, Jaber MA, Al-Ansari MM, Sumaily KM, Al-Alwan M, Sabi EM, Malkawi AK, Abdel Rahman AM. Metabolic Alteration of MCF-7 Cells upon Indirect Exposure to E. coli Secretome: A Model of Studying the Microbiota Effect on Human Breast Tissue. Metabolites 2023; 13:938. [PMID: 37623881 PMCID: PMC10456566 DOI: 10.3390/metabo13080938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
According to studies, the microbiome may contribute to the emergence and spread of breast cancer. E. coli is one of the Enterobacteriaceae family recently found to be present as part of the breast tissue microbiota. In this study, we focused on the effect of E. coli secretome free of cells on MCF-7 metabolism. Liquid chromatography-mass spectrometry (LC-MS) metabolomics was used to study the E. coli secretome and its role in MCF-7 intra- and extracellular metabolites. A comparison was made between secretome-exposed cells and unexposed controls. Our analysis revealed significant alterations in 31 intracellular and 55 extracellular metabolites following secretome exposure. Several metabolic pathways, including lactate, aminoacyl-tRNA biosynthesis, purine metabolism, and energy metabolism, were found to be dysregulated upon E. coli secretome exposure. E. coli can alter the breast cancer cells' metabolism through its secretome which disrupts key metabolic pathways of MCF-7 cells. These microbial metabolites from the secretome hold promise as biomarkers of drug resistance or innovative approaches for cancer treatment, either as standalone therapies or in combination with other medicines.
Collapse
Affiliation(s)
- Reem H. AlMalki
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Malak A. Jaber
- Pharmaceutical Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan;
| | - Mysoon M. Al-Ansari
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Khalid M. Sumaily
- Clinical Biochemistry Unit, Pathology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.M.S.); (E.M.S.)
| | - Monther Al-Alwan
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia;
| | - Essa M. Sabi
- Clinical Biochemistry Unit, Pathology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.M.S.); (E.M.S.)
| | - Abeer K. Malkawi
- Department of Chemistry and Biochemistry, Université Du Québec à Montréal, Montréal, QC H3C 3P8, Canada;
| | - Anas M. Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia
| |
Collapse
|
48
|
Ujlaki G, Kovács T, Vida A, Kókai E, Rauch B, Schwarcz S, Mikó E, Janka E, Sipos A, Hegedűs C, Uray K, Nagy P, Bai P. Identification of Bacterial Metabolites Modulating Breast Cancer Cell Proliferation and Epithelial-Mesenchymal Transition. Molecules 2023; 28:5898. [PMID: 37570868 PMCID: PMC10420980 DOI: 10.3390/molecules28155898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer patients are characterized by the oncobiotic transformation of multiple microbiome communities, including the gut microbiome. Oncobiotic transformation of the gut microbiome impairs the production of antineoplastic bacterial metabolites. The goal of this study was to identify bacterial metabolites with antineoplastic properties. We constructed a 30-member bacterial metabolite library and screened the library compounds for effects on cell proliferation and epithelial-mesenchymal transition. The metabolites were applied to 4T1 murine breast cancer cells in concentrations corresponding to the reference serum concentrations. However, yric acid, glycolic acid, d-mannitol, 2,3-butanediol, and trans-ferulic acid exerted cytostatic effects, and 3-hydroxyphenylacetic acid, 4-hydroxybenzoic acid, and vanillic acid exerted hyperproliferative effects. Furthermore, 3-hydroxyphenylacetic acid, 4-hydroxybenzoic acid, 2,3-butanediol, and hydrocinnamic acid inhibited epithelial-to-mesenchymal (EMT) transition. We identified redox sets among the metabolites (d-mannitol-d-mannose, 1-butanol-butyric acid, ethylene glycol-glycolic acid-oxalic acid), wherein only one partner within the set (d-mannitol, butyric acid, glycolic acid) possessed bioactivity in our system, suggesting that changes to the local redox potential may affect the bacterial secretome. Of the nine bioactive metabolites, 2,3-butanediol was the only compound with both cytostatic and anti-EMT properties.
Collapse
Affiliation(s)
- Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - András Vida
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Endre Kókai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Boglára Rauch
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Szandra Schwarcz
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Eszter Janka
- Department of Dermatology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
| | - Péter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Peter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.U.); (T.K.); (A.V.); (E.K.); (B.R.); (S.S.); (E.M.); (A.S.); (C.H.); (K.U.)
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Biology and Signaling Research Group ELKH, 4032 Debrecen, Hungary
| |
Collapse
|
49
|
Gong Y, Huang X, Wang M, Liang X. Intratumor microbiota: a novel tumor component. J Cancer Res Clin Oncol 2023; 149:6675-6691. [PMID: 36639531 DOI: 10.1007/s00432-023-04576-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Bacteria have been found in tumors for over 100 years, but the irreproducibility of experiments on bacteria, the limitations of science and technology, and the contamination of the host environment have severely hampered most research into the role of bacteria in carcinogenesis and cancer treatment. With the development of molecular tools and techniques (e.g., macrogenomics, metabolomics, lipidomics, and macrotranscriptomics), the complex relationships between hosts and different microorganisms are gradually being deciphered. In the past, attention has been focused on the impact of the gut microbiota, the site where the body's microbes gather most, on tumors. However, little is known about the role of microbes from other sites, particularly the intratumor microbiota, in cancer. In recent years, an increasing number of studies have identified the presence of symbiotic microbiota within a large number of tumors, bringing the intratumor microbiota into the limelight. In this review, we aim to provide a better understanding of the role of the intratumor microbiota in cancer, to provide direction for future experimental and translational research, and to offer new approaches to the treatment of cancer and the improvement of patient prognosis.
Collapse
Affiliation(s)
- Yanyu Gong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xinqi Huang
- Excellent Class, Clinical Medicine, Grade 20, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Minhui Wang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoqiu Liang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
50
|
Bernardo G, Le Noci V, Di Modica M, Montanari E, Triulzi T, Pupa SM, Tagliabue E, Sommariva M, Sfondrini L. The Emerging Role of the Microbiota in Breast Cancer Progression. Cells 2023; 12:1945. [PMID: 37566024 PMCID: PMC10417285 DOI: 10.3390/cells12151945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Emerging evidence suggests a profound association between the microbiota composition in the gastrointestinal tract and breast cancer progression. The gut microbiota plays a crucial role in modulating the immune response, releasing metabolites, and modulating estrogen levels, all of which have implications for breast cancer growth. However, recent research has unveiled a novel aspect of the relationship between the microbiota and breast cancer, focusing on microbes residing within the mammary tissue, which was once considered sterile. These localized microbial communities have been found to change in the presence of a tumor as compared to healthy mammary tissue, unraveling their potential contribution to tumor progression. Studies have identified specific bacterial species that are enriched within breast tumors and have highlighted the mechanisms by which even these microbes influence cancer progression through immune modulation, direct carcinogenic activity, and effects on cellular pathways involved in cell proliferation or apoptosis. This review aims to provide an overview of the current knowledge on the mechanisms of crosstalk between the gut/mammary microbiota and breast cancer. Understanding this intricate interplay holds promise for developing innovative therapeutic approaches.
Collapse
Affiliation(s)
- Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (V.L.N.); (M.S.)
| | - Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (V.L.N.); (M.S.)
| | - Martina Di Modica
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| | - Elena Montanari
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| | - Tiziana Triulzi
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| | - Serenella M. Pupa
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| | - Elda Tagliabue
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (V.L.N.); (M.S.)
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (G.B.); (V.L.N.); (M.S.)
- Microenvironment and Biomarkers of Solid Tumors, Experimental Oncology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, 20133 Milan, Italy; (M.D.M.); (E.M.); (T.T.); (S.M.P.); (E.T.)
| |
Collapse
|