1
|
Roesel R, Strati F, Basso C, Epistolio S, Spina P, Djordjevic J, Sorrenti E, Villa M, Cianfarani A, Mongelli F, Galafassi J, Popeskou SG, Facciotti F, Caprera C, Melle F, Majno-Hurst PE, Franzetti-Pellanda A, De Dosso S, Bonfiglio F, Frattini M, Christoforidis D, Iezzi G. Combined tumor-associated microbiome and immune gene expression profiling predict response to neoadjuvant chemo-radiotherapy in locally advanced rectal cancer. Oncoimmunology 2025; 14:2465015. [PMID: 39992705 PMCID: PMC11853554 DOI: 10.1080/2162402x.2025.2465015] [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: 03/04/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Locally advanced rectal cancer (LARC) is treated with neoadjuvant chemo-radiotherapy (nCRT) followed by surgery. A minority of patients show complete response (CR) to nCRT and may avoid surgery and its functional consequences. Instead, most patients show non-complete response (non-CR) and may benefit from additional treatments to increase CR rates. Reliable predictive markers are lacking. Aim of this study was to identify novel signatures predicting nCRT responsiveness. We performed a combined analysis of tumor-associated microbiome and immune gene expression profiling of diagnostic biopsies from 70 patients undergoing nCRT followed by rectal resection, including 16 with CR and 54 with non-CR. Findings were validated by an independent cohort of 49 patients, including 7 with CR and 42 with non-CR. Intratumoral microbiota significantly differed between CR and non-CR groups at genus and species level. Colonization by bacterial species of Ruminococcus genera was consistently associated with CR, whereas abundance of Fusobacterium, Porhpyromonas, and Oscillibacter species predicted non-CR. Immune gene profiling revealed a panel of 59 differentially expressed genes and significant upregulation of IFN-gamma and -alpha response in patients with CR. Integrated microbiome and immune gene profiling analysis unraveled clustering of microbial taxa with each other and with immune cell-related genes and allowed the identification of a combined signature correctly identifying non-CRS in both cohorts. Thus, combined intratumoral microbiome-immune profiling improves the prediction of response to nCRT. Correct identification of unresponsive patients and of bacteria promoting responsiveness might lead to innovative therapeutic approaches based on gut microbiota pre-conditioning to increase nCRT effectiveness in LARC.
Collapse
Affiliation(s)
- Raffaello Roesel
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Camilla Basso
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Samantha Epistolio
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Paolo Spina
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Julija Djordjevic
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Elisa Sorrenti
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Martina Villa
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Agnese Cianfarani
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Francesco Mongelli
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Jacopo Galafassi
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Sotirios G. Popeskou
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Federica Facciotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Cecilia Caprera
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Melle
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Pietro Edoardo Majno-Hurst
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | | | - Sara De Dosso
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Ferdinando Bonfiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, Naples, Italy
- CEINGE Advanced Biotechnology Franco Salvatore, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Milo Frattini
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Dimitrios Christoforidis
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Visceral Surgery, CHUV, University of Lausanne, Lausanne, Switzerland
| | - Giandomenica Iezzi
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
2
|
Emoto S, Inoue R, Murai S, Inagaki Y, Nozawa H, Sasaki K, Murono K, Kaneko K, Yokoyama Y, Abe S, Nagai Y, Shinagawa T, Tachikawa Y, Okada S, Tsukahara T, Ohashi K, Ohno M, Andoh A, Ishihara S. A pilot study of gut mucosal and faecal microbiota in rectal cancer: associations with histological response and adverse events following preoperative chemoradiotherapy. Colorectal Dis 2025; 27:e70106. [PMID: 40312795 PMCID: PMC12046106 DOI: 10.1111/codi.70106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
AIM Preoperative chemoradiotherapy (CRT) is administered for locally advanced rectal cancer (LARC); however, its efficacy and toxicity vary among patients. This study aimed to elucidate the relationship between the gut microbiota and the effectiveness and adverse events of CRT. METHODS This prospective study included 21 patients with LARC with no history of antibiotic or probiotic administration for 6 months. Tumour mucosa, non-tumour mucosa and faecal samples were collected before and after CRT, and bacterial DNA was extracted. Metataxonomic analysis targeting the V3 and V4 regions of the 16S rRNA gene was conducted to determine the diversity and composition of the microbiota. Linear discriminant analysis effect size (LEfSe) was used to explore potential bacterial taxa predicting pathological complete response (pCR) and treatment-associated diarrhoea, which are major adverse events of CRT. RESULTS Among the 21 patients, five achieved pCR and seven experienced severe treatment-associated diarrhoea. There were no significant differences in α-diversity and β-diversity of the microbiota between the groups at any sampling sites before or after CRT. Exploratory analysis using LEfSe identified Peptostreptococcus, Coprococcus and Phoceaicola in the tumour mucosa before CRT as significant indicators for achieving pCR. Additionally, Collinsella, Haemophilus and Desulfovibrionaceae are associated with treatment-associated diarrhoea. Microbiome composition changed before and after CRT, with a notable decrease in the genus Fusobacterium_C and other taxa. β-diversity in the tumour area also changed significantly (P = 0.03). CONCLUSIONS This study suggests an association between the gut microbiota, the therapeutic effectiveness of CRT and the occurrence of treatment-associated diarrhoea in rectal cancer. These results indicate the potential for predicting treatment efficacy and adverse events based on the microbiota composition.
Collapse
Affiliation(s)
- Shigenobu Emoto
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Ryo Inoue
- Laboratory of Animal ScienceSetsunan UniversityHirakataJapan
| | - Shin Murai
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Yuriko Inagaki
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Hiroaki Nozawa
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Kazuhito Sasaki
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Koji Murono
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Kensuke Kaneko
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | | | - Shinya Abe
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | - Yuzo Nagai
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | | | | | - Satoshi Okada
- Department of Surgical OncologyUniversity of TokyoTokyoJapan
| | | | - Kai Ohashi
- Kyoto Institute of Nutrition and PathologyUjitawaraJapan
| | - Masashi Ohno
- Department of GastroenterologyShiga University of Medical ScienceOtsuJapan
| | - Akira Andoh
- Department of GastroenterologyShiga University of Medical ScienceOtsuJapan
| | | |
Collapse
|
3
|
Greco L, Rubbino F, Ferrari C, Cameletti M, Grizzi F, Bonelli F, Malesci A, Mazzone M, Ricciardiello L, Laghi L. Association of Fusobacterium nucleatum with colorectal cancer molecular subtypes and its outcome: a systematic review. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2025; 6:e5. [PMID: 40297307 PMCID: PMC12035788 DOI: 10.1017/gmb.2025.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
Colorectal cancer (CRC) represents a relevant public health problem, with high incidence and mortality in Western countries. CRC can occur as sporadic (65%-75%), common familial (25%), or as a consequence of an inherited predisposition (up to 10%). While unravelling its genetic basis has been a long trip leading to relevant clinical implementation over more than 30 years, other contributing factors remain to be clarified. Among these, micro-organisms have emerged as critical players in the development and progression of the disease, as well as for CRC treatment response. Fusobacterium nucleatum (Fn) has been associated with CRC development in both pre-clinical models and clinical settings. Fusobacteria are core members of the human oral microbiome, while being less prevalent in the healthy gut, prompting questions about their localization in CRC and its precursor lesions. This review aims to critically discuss the evidence connecting Fn with CRC pathogenesis, its molecular subtypes and clinical outcomes.
Collapse
Affiliation(s)
- Luana Greco
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Federica Rubbino
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Clarissa Ferrari
- Research and Clinical Trials Office, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | | | - Fabio Grizzi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
| | | | | | - Massimiliano Mazzone
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Macrophage Dynamics Lab, IRCCS Humanitas Research Hospital, Milan, Italy
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Luigi Ricciardiello
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
4
|
Zhao Z, Peng Y, Yang Y, Li S, Ling J, Zhu Z, He C. ATP13A2 as a prognostic biomarker and its correlation with immune infiltration in cervical cancer: A retrospective study. J Cell Mol Med 2025; 29:e70097. [PMID: 40197818 PMCID: PMC11976316 DOI: 10.1111/jcmm.70097] [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: 02/20/2024] [Revised: 06/11/2024] [Accepted: 09/03/2024] [Indexed: 04/10/2025] Open
Abstract
While the oncogene ATP13A2 is reportedly involved in colorectal cancer, its role in cervical cancer (CC) has yet to be fully characterized. In this study, we investigated ATP13A2 as a potential prognostic biomarker of CC. To this end, we compared CC tissues with normal tissues to identify differentially expressed genes, identifying ATP13A2 as a potential marker of CC. Elevated ATP13A2 expression levels were identified in CC samples compared to noncancerous samples across various data sets, with further immunohistochemical validation. Functional enrichment analysis revealed that ATP13A2 plays an essential role in the CXCL12-activated CXCR4 signalling pathway and chemotaxis regulation, which may alter immune infiltration. Notably, increased ATP13A2 levels were associated with poor overall survival. Furthermore, multiple clinical characteristics were significantly associated with ATP13A2 expression. Additionally, tumour bacterial infiltration was assessed using weighted co-expression network analysis, revealing a relationship between ATP13A2 expression and bacteria in the CC tumour microenvironment. Our results suggest that ATP13A2 is a promising diagnostic and prognostic marker for CC. However, further large-scale studies are needed to fully elucidate the mechanisms underlying the involvement of ATP13A2 in CC.
Collapse
Affiliation(s)
- Zhi Zhao
- Zhengzhou Yihe Hospital, Postdoctoral Innovation Practice BaseHenan UniversityZhengzhouHenanChina
- Department of Breast SurgeryGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Yijie Peng
- Department of Hepatobiliary and Pancreatic SurgeryThe Central Hospital of ShaoyangShaoyangHunanChina
| | - Yuanyuan Yang
- Clinical Research Center for Women's Reproductive Health in Hunan ProvinceChangshaHunanChina
- Reproductive Medicine CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Shuaiyu Li
- School of Information ScienceKyushu UniversityFukuokaJapan
| | - Jiang Ling
- Department of Forensic Science, School of Basic Medical SciencesCentral South UniversityChangshaHunanChina
| | - Zhenyu Zhu
- Department of Breast SurgeryGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Chenfeng He
- Department of Integrative BioanalyticsInstitute of Development, Aging and Cancer (IDAC), Tohoku UniversitySendaiJapan
| |
Collapse
|
5
|
Zhao F, An R, Ma Y, Yu S, Gao Y, Wang Y, Yu H, Xie X, Zhang J. Integrated spatial multi-omics profiling of Fusobacterium nucleatum in breast cancer unveils its role in tumour microenvironment modulation and cancer progression. Clin Transl Med 2025; 15:e70273. [PMID: 40070022 PMCID: PMC11897063 DOI: 10.1002/ctm2.70273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
Tumour-associated microbiota are integral components of the tumour microenvironment (TME). However, previous studies on intratumoral microbiota primarily rely on bulk tissue analysis, which may obscure their spatial distribution and localized effects. In this study, we applied in situ spatial-profiling technology to investigate the spatial distribution of intratumoral microbiota in breast cancer and their interactions with the local TME. Using 5R 16S rRNA gene sequencing and RNAscope FISH/CISH on patients' tissue, we identified significant spatial heterogeneity in intratumoral microbiota, with Fusobacterium nucleatum (F. nucleatum) predominantly localized in tumour cell-rich areas. GeoMx digital spatial profiling (DSP) revealed that regions colonized by F. nucleatum exhibit significant influence on the expression of RNAs and proteins involved in proliferation, migration and invasion. In vitro studies indicated that co-culture with F. nucleatum significantly stimulates the proliferation and migration of breast cancer cells. Integrative spatial multi-omics and co-culture transcriptomic analyses highlighted the MAPK signalling pathways as key altered pathways. By intersecting these datasets, VEGFD and PAK1 emerged as critical upregulated proteins in F. nucleatum-positive regions, showing strong positive correlations with MAPK pathway proteins. Moreover, the upregulation of VEGFD and PAK1 by F. nucleatum was confirmed in co-culture experiments, and their knockdown significantly reduced F. nucleatum-induced proliferation and migration. In conclusion, intratumoral microbiota in breast cancer exhibit significant spatial heterogeneity, with F. nucleatum colonization markedly altering tumour cell protein expression to promote progression and migration. These findings provide novel perspectives on the role of microbiota in breast cancer, identify potential therapeutic targets, and lay the foundation for future cancer treatments. KEY POINTS: Intratumoral Fusobacterium nucleatum exhibits significant spatial heterogeneity within breast cancer tissues. F. nucleatum colonization alters the expression of key proteins involved in tumour progression and migration. The MAPK signalling pathway is a critical mediator of F. nucleatum-induced breast cancer cell proliferation and migration. VEGFD and PAK1 are potential therapeutic targets to mitigate F. nucleatum-induced tumour progression.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Rui An
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yilei Ma
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Shaobo Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yuzhen Gao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yanzhong Wang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Haitao Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Xinyou Xie
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Jun Zhang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| |
Collapse
|
6
|
Zalila-Kolsi I, Dhieb D, Osman HA, Mekideche H. The Gut Microbiota and Colorectal Cancer: Understanding the Link and Exploring Therapeutic Interventions. BIOLOGY 2025; 14:251. [PMID: 40136508 PMCID: PMC11939563 DOI: 10.3390/biology14030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
CRC remains a significant public health challenge due to its high prevalence and mortality rates. Emerging evidence highlights the critical role of the gut microbiota in both the pathogenesis of CRC and the efficacy of treatment strategies, including chemotherapy and immunotherapy. Dysbiosis, characterized by imbalances in microbial communities, has been implicated in CRC progression and therapeutic outcomes. This review examines the intricate relationship between gut microbiota composition and CRC, emphasizing the potential for microbial profiles to serve as biomarkers for early detection and prognosis. Various interventions, such as prebiotics, probiotics, postbiotics, fecal microbiota transplantation, and dietary modifications, aim to restore microbiota balance and shift dysbiosis toward eubiosis, thereby improving health outcomes. Additionally, the integration of microbial profiling into clinical practice could enhance diagnostic capabilities and personalize treatment strategies, advancing the field of oncology. The study of intratumoral microbiota offers new diagnostic and prognostic tools that, combined with artificial intelligence algorithms, could predict treatment responses and assess the risk of adverse effects. Given the growing understanding of the gut microbiome-cancer axis, developing microbiota-oriented strategies for CRC prevention and treatment holds promise for improving patient care and clinical outcomes.
Collapse
Affiliation(s)
- Imen Zalila-Kolsi
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Dhoha Dhieb
- College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussam A. Osman
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Hadjer Mekideche
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| |
Collapse
|
7
|
Wang N, Wu S, Huang L, Hu Y, He X, He J, Hu B, Xu Y, Rong Y, Yuan C, Zeng X, Wang F. Intratumoral microbiome: implications for immune modulation and innovative therapeutic strategies in cancer. J Biomed Sci 2025; 32:23. [PMID: 39966840 PMCID: PMC11837407 DOI: 10.1186/s12929-025-01117-x] [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: 09/09/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Recent advancements have revealed the presence of a microbiome within tumor tissues, underscoring the crucial role of the tumor microbiome in the tumor ecosystem. This review delves into the characteristics of the intratumoral microbiome, underscoring its dual role in modulating immune responses and its potential to both suppress and promote tumor growth. We examine state-of-the-art techniques for detecting and analyzing intratumoral bacteria, with a particular focus on their interactions with the immune system and the resulting implications for cancer prognosis and treatment. By elucidating the intricate crosstalk between the intratumoral microbiome and the host immune system, we aim to uncover novel therapeutic strategies that enhance the efficacy of cancer treatments. Additionally, this review addresses the existing challenges and future prospects within this burgeoning field, advocating for the integration of microbiome research into comprehensive cancer therapy frameworks.
Collapse
Affiliation(s)
- Na Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Si Wu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lanxiang Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yue Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xin He
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jourong He
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ben Hu
- Center for Tumor Precision Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yaqi Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuan Rong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China.
| | - Xiantao Zeng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
8
|
Liang L, Kong C, Li J, Liu G, Wei J, Wang G, Wang Q, Yang Y, Shi D, Li X, Ma Y. Distinct microbes, metabolites, and the host genome define the multi-omics profiles in right-sided and left-sided colon cancer. MICROBIOME 2024; 12:274. [PMID: 39731152 DOI: 10.1186/s40168-024-01987-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Studies have reported clinical heterogeneity between right-sided colon cancer (RCC) and left-sided colon cancer (LCC). However, none of these studies used multi-omics analysis combining genetic regulation, microbiota, and metabolites to explain the site-specific difference. METHODS Here, 494 participants from a 16S rRNA gene sequencing cohort (50 RCC, 114 LCC, and 100 healthy controls) and a multi-omics cohort (63 RCC, 79 LCC, and 88 healthy controls) were analyzed. 16S rRNA gene, metagenomic sequencing, and metabolomics analyses of fecal samples were evaluated to identify tumor location-related bacteria and metabolites. Whole-exome sequencing (WES) and transcriptome sequencing (RNA-seq) were conducted to obtain the mutation burden and genomic expression pattern. RESULTS We found unique profiles of the intestinal microbiome, metabolome, and host genome between RCC and LCC. The bacteria Flavonifractor plautii (Fp) and Fusobacterium nucleatum, the metabolites L-phenylalanine, and the host genes PHLDA1 and WBP1 were the key omics features of RCC; whereas the bacteria Bacteroides sp. A1C1 (B.A1C1) and Parvimonas micra, the metabolites L-citrulline and D-ornithine, and the host genes TCF25 and HLA-DRB5 were considered the dominant omics features in LCC. Multi-omics correlation analysis indicated that RCC-enriched Fp was related to the accumulation of the metabolite L-phenylalanine and the suppressed WBP1 signal in RCC patients. In addition, LCC-enriched B.A1C1 was associated with the accumulation of the metabolites D-ornithine and L-citrulline as well as activation of the genes TCF25, HLA-DRB5, and AC079354.1. CONCLUSION Our findings identify previously unknown links between intestinal microbiota alterations, metabolites, and host genomics in RCC vs. LCC, suggesting that it may be possible to treat colorectal cancer (CRC) by targeting the gut microbiota-host interaction. Video Abstract.
Collapse
Affiliation(s)
- Lei Liang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Kong
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinming Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guang Liu
- Guangdong Hongyuan Pukang Medical Technology Co., Ltd., Guangdong, China
| | - Jinwang Wei
- GenomiCare Biotechnology Co. Ltd., Shanghai, China
| | - Guan Wang
- GenomiCare Biotechnology Co. Ltd., Shanghai, China
| | - Qinying Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Debing Shi
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Kunath BJ, De Rudder C, Laczny CC, Letellier E, Wilmes P. The oral-gut microbiome axis in health and disease. Nat Rev Microbiol 2024; 22:791-805. [PMID: 39039286 DOI: 10.1038/s41579-024-01075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
The human body hosts trillions of microorganisms throughout many diverse habitats with different physico-chemical characteristics. Among them, the oral cavity and the gut harbour some of the most dense and diverse microbial communities. Although these two sites are physiologically distinct, they are directly connected and can influence each other in several ways. For example, oral microorganisms can reach and colonize the gastrointestinal tract, particularly in the context of gut dysbiosis. However, the mechanisms of colonization and the role that the oral microbiome plays in causing or exacerbating diseases in other organs have not yet been fully elucidated. Here, we describe recent advances in our understanding of how the oral and intestinal microbiota interplay in relation to their impact on human health and disease.
Collapse
Affiliation(s)
- Benoit J Kunath
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
10
|
Dorobisz K, Dorobisz T, Pazdro-Zastawny K, Czyż K, Janczak M. The Influence of the Microbiome on the Complications of Radiotherapy and Its Effectiveness in Patients with Laryngeal Cancer. Cancers (Basel) 2024; 16:3707. [PMID: 39518144 PMCID: PMC11545705 DOI: 10.3390/cancers16213707] [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: 10/20/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Radiotherapy is an effective method of treating cancer and affects 50% of patients. Intensity-modulated radiotherapy (IMRT) is a modernized method of classical radiation used in the treatment of laryngeal cancer. Treatment with intent to preserve the larynx is not always safe or complication-free. The microbiome may significantly influence the effectiveness of oncological treatment, especially radiotherapy, and may also be modified by the toxic response to radiation. OBJECTIVE The aim of the study was to prospectively assess the microbiome and its influence on radiotherapy toxicity in patients with laryngeal cancer. RESULTS Statistically significant risk factors for complications after radiotherapy were the percentage of Porphyromonas of at least 6.7%, the percentage of Fusobacterium of at least 2.6% and the percentage of Catonella of at least 2.6%. CONCLUSIONS The importance of the microbiome in oncology has been confirmed in many studies. Effective radiotherapy treatment and the prevention of radiation-induced oral mucositis is a challenge in oncology. The microbiome may be an important part of personalized cancer treatment. The assessment of the microbiome of patients diagnosed with cancer may provide the opportunity to predict the response to treatment and its effectiveness. The influence of the microbiome may be important in predicting the risk group for radiotherapy treatment failure. The possibility of modifying the microbiome may become a goal to improve the prognosis of patients with laryngeal cancer. Fusobacterium, Porphyromonas and Catonella are important risk factors for radiation-induced oral mucositis in patients with laryngeal cancer.
Collapse
Affiliation(s)
- Karolina Dorobisz
- Department of Otolaryngology, Head and Neck Surgery, Wrocław Medical University, 50-367 Wroclaw, Poland
| | - Tadeusz Dorobisz
- Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Katarzyna Pazdro-Zastawny
- Department of Otolaryngology, Head and Neck Surgery, Wrocław Medical University, 50-367 Wroclaw, Poland
| | - Katarzyna Czyż
- Institute of Animal Breeding, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Marzena Janczak
- Institute of Animal Breeding, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| |
Collapse
|
11
|
González A, Badiola I, Fullaondo A, Rodríguez J, Odriozola A. Personalised medicine based on host genetics and microbiota applied to colorectal cancer. ADVANCES IN GENETICS 2024; 112:411-485. [PMID: 39396842 DOI: 10.1016/bs.adgen.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) ranks second in incidence and third in cancer mortality worldwide. This situation, together with the understanding of the heterogeneity of the disease, has highlighted the need to develop a more individualised approach to its prevention, diagnosis and treatment through personalised medicine. This approach aims to stratify patients according to risk, predict disease progression and determine the most appropriate treatment. It is essential to identify patients who may respond adequately to treatment and those who may be resistant to treatment to avoid unnecessary therapies and minimise adverse side effects. Current research is focused on identifying biomarkers such as specific mutated genes, the type of mutations and molecular profiles critical for the individualisation of CRC diagnosis, prognosis and treatment guidance. In addition, the study of the intestinal microbiota as biomarkers is being incorporated due to the growing scientific evidence supporting its influence on this disease. This article comprehensively addresses the use of current and emerging diagnostic, prognostic and predictive biomarkers in precision medicine against CRC. The effects of host genetics and gut microbiota composition on new approaches to treating this disease are discussed. How the gut microbiota could mitigate the side effects of treatment is reviewed. In addition, strategies to modulate the gut microbiota, such as dietary interventions, antibiotics, and transplantation of faecal microbiota and phages, are discussed to improve CRC prevention and treatment. These findings provide a solid foundation for future research and improving the care of CRC patients.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | | | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
12
|
Cao C, Yue S, Lu A, Liang C. Host-Gut Microbiota Metabolic Interactions and Their Role in Precision Diagnosis and Treatment of Gastrointestinal Cancers. Pharmacol Res 2024; 207:107321. [PMID: 39038631 DOI: 10.1016/j.phrs.2024.107321] [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: 02/24/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
The critical role of the gut microbiome in gastrointestinal cancers is becoming increasingly clear. Imbalances in the gut microbial community, referred to as dysbiosis, are linked to increased risks for various forms of gastrointestinal cancers. Pathogens like Fusobacterium and Helicobacter pylori relate to the onset of esophageal and gastric cancers, respectively, while microbes such as Porphyromonas gingivalis and Clostridium species have been associated with a higher risk of pancreatic cancer. In colorectal cancer, bacteria such as Fusobacterium nucleatum are known to stimulate the growth of tumor cells and trigger cancer-promoting pathways. On the other hand, beneficial microbes like Bifidobacteria offer a protective effect, potentially inhibiting the development of gastrointestinal cancers. The potential for therapeutic interventions that manipulate the gut microbiome is substantial, including strategies to engineer anti-tumor metabolites and employ microbiota-based treatments. Despite the progress in understanding the influence of the microbiome on gastrointestinal cancers, significant challenges remain in identifying and understanding the precise contributions of specific microbial species and their metabolic products. This knowledge is essential for leveraging the role of the gut microbiome in the development of precise diagnostics and targeted therapies for gastrointestinal cancers.
Collapse
Affiliation(s)
- Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Siran Yue
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510006, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| |
Collapse
|
13
|
Bukavina L, Ginwala R, Eltoukhi M, Sindhani M, Prunty M, Geynisman DM, Ghatalia P, Valentine H, Calaway A, Correa AF, Brown JR, Mishra K, Plimack ER, Kutikov A, Ghannoum M, Elshaer M, Retuerto M, Ponsky L, Uzzo RG, Abbosh PH. Role of Gut Microbiome in Neoadjuvant Chemotherapy Response in Urothelial Carcinoma: A Multi-institutional Prospective Cohort Evaluation. CANCER RESEARCH COMMUNICATIONS 2024; 4:1505-1516. [PMID: 38747616 PMCID: PMC11181990 DOI: 10.1158/2767-9764.crc-23-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/21/2024] [Accepted: 05/09/2024] [Indexed: 05/28/2024]
Abstract
Neoadjuvant chemotherapy (NAC) is linked with clinical advantages in urothelial carcinoma for patients with muscle-invasive bladder cancer (MIBC). Despite comprehensive research into the influence of tumor mutation expression profiles and clinicopathologic factors on chemotherapy response, the role of the gut microbiome (GM) in bladder cancer chemotherapy response remains poorly understood. This study examines the variance in the GM of patients with bladder cancer compared with healthy adults, and investigates GM compositional differences between patients who respond to chemotherapy versus those who exhibit residual disease.Our study reveals distinct clustering, effectively separating the bladder cancer and healthy cohorts. However, no significant differences were observed between chemotherapy responders and nonresponders within community subgroups. Machine learning models based on responder status outperformed clinical variables in predicting complete response (AUC 0.88 vs. AUC 0.50), although no single microbial species emerged as a fully reliable biomarker.The evaluation of short chain fatty acid (SCFA) concentration in blood and stool revealed no correlation with responder status. Still, SCFA analysis showed a higher abundance of Akkermansia (rs = 0.51, P = 0.017) and Clostridia (rs = 0.52, P = 0.018), which correlated with increased levels of detectable fecal isobutyric acid. Higher levels of fecal Lactobacillus (rs = 0.49, P = 0.02) and Enterobacteriaceae (rs = 0.52, P < 0.03) correlated with increased fecal propionic acid.In conclusion, our study constitutes the first large-scale, multicenter assessment of GM composition, suggesting the potential for a complex microbial signature to predict patients more likely to respond to NAC based on multiple taxa. SIGNIFICANCE Our study highlights results that link the composition of the GM to the efficacy of NAC in MIBC. We discovered that patients with higher levels of Bacteroides experienced a worse response to NAC. This microbial signature shows promise as a superior predictor of treatment response over traditional clinical variables. Although preliminary, our findings advocate for larger, more detailed studies to validate these associations.
Collapse
Affiliation(s)
- Laura Bukavina
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Cleveland Clinic Glickman Urologic Institute, Cleveland, Ohio
- Case Western Reserve School of Medicine, Cleveland, Ohio
| | | | | | | | - Megan Prunty
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | | | | | - Adam Calaway
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Jason R. Brown
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Kirtishri Mishra
- Case Western Reserve School of Medicine, Cleveland, Ohio
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | | | | | | | | | - Lee Ponsky
- Case Western Reserve School of Medicine, Cleveland, Ohio
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Philip H. Abbosh
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Albert Einstein Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Messaritakis I, Koulouris A, Boukla E, Vogiatzoglou K, Lagkouvardos I, Intze E, Sfakianaki M, Chondrozoumaki M, Karagianni M, Athanasakis E, Xynos E, Tsiaoussis J, Christodoulakis M, Flamourakis ME, Tsagkataki ES, Giannikaki L, Chliara E, Mavroudis D, Tzardi M, Souglakos J. Exploring Gut Microbiome Composition and Circulating Microbial DNA Fragments in Patients with Stage II/III Colorectal Cancer: A Comprehensive Analysis. Cancers (Basel) 2024; 16:1923. [PMID: 38792001 PMCID: PMC11119035 DOI: 10.3390/cancers16101923] [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: 03/31/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) significantly contributes to cancer-related mortality, necessitating the exploration of prognostic factors beyond TNM staging. This study investigates the composition of the gut microbiome and microbial DNA fragments in stage II/III CRC. METHODS A cohort of 142 patients with stage II/III CRC and 91 healthy controls underwent comprehensive microbiome analysis. Fecal samples were collected for 16S rRNA sequencing, and blood samples were tested for the presence of microbial DNA fragments. De novo clustering analysis categorized individuals based on their microbial profiles. Alpha and beta diversity metrics were calculated, and taxonomic profiling was conducted. RESULTS Patients with CRC exhibited distinct microbial composition compared to controls. Beta diversity analysis confirmed CRC-specific microbial profiles. Taxonomic profiling revealed unique taxonomies in the patient cohort. De novo clustering separated individuals into distinct groups, with specific microbial DNA fragment detection associated with certain patient clusters. CONCLUSIONS The gut microbiota can differentiate patients with CRC from healthy individuals. Detecting microbial DNA fragments in the bloodstream may be linked to CRC prognosis. These findings suggest that the gut microbiome could serve as a prognostic factor in stage II/III CRC. Identifying specific microbial markers associated with CRC prognosis has potential clinical implications, including personalized treatment strategies and reduced healthcare costs. Further research is needed to validate these findings and uncover underlying mechanisms.
Collapse
Affiliation(s)
- Ippokratis Messaritakis
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Andreas Koulouris
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Eleni Boukla
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Konstantinos Vogiatzoglou
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Ilias Lagkouvardos
- Department of Clinical Microbiology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (I.L.); (E.I.)
| | - Evangelia Intze
- Department of Clinical Microbiology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (I.L.); (E.I.)
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Maria Chondrozoumaki
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Michaela Karagianni
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
| | - Elias Athanasakis
- Department of General Surgery, Heraklion University Hospital, 71100 Heraklion, Greece;
| | - Evangelos Xynos
- Department of Surgery, Creta Interclinic Hospital of Heraklion, 71305 Heraklion, Greece
| | - John Tsiaoussis
- Department of Anatomy, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | | | | | - Eleni S. Tsagkataki
- Department of General Surgery, Venizeleio General Hospital, 71409 Heraklion, Greece (M.E.F.)
| | - Linda Giannikaki
- Histopathology, Venizeleio General Hospital, 71409 Heraklion, Greece
| | - Evdoxia Chliara
- Histopathology, Venizeleio General Hospital, 71409 Heraklion, Greece
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
- Department of Medical Oncology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Maria Tzardi
- Laboratory of Pathology, University General Hospital of Heraklion, 70013 Heraklion, Greece;
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (A.K.); (M.C.); (D.M.)
- Department of Medical Oncology, University Hospital of Heraklion, 71110 Heraklion, Greece
| |
Collapse
|
15
|
Udayasuryan B, Zhou Z, Ahmad RN, Sobol P, Deng C, Nguyen TTD, Kodikalla S, Morrison R, Goswami I, Slade DJ, Verbridge SS, Lu C. Fusobacterium nucleatum infection modulates the transcriptome and epigenome of HCT116 colorectal cancer cells in an oxygen-dependent manner. Commun Biol 2024; 7:551. [PMID: 38720110 PMCID: PMC11079022 DOI: 10.1038/s42003-024-06201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Fusobacterium nucleatum, a gram-negative oral bacterium, has been consistently validated as a strong contributor to the progression of several types of cancer, including colorectal (CRC) and pancreatic cancer. While previous in vitro studies have shown that intracellular F. nucleatum enhances malignant phenotypes such as cell migration, the dependence of this regulation on features of the tumor microenvironment (TME) such as oxygen levels are wholly uncharacterized. Here we examine the influence of hypoxia in facilitating F. nucleatum invasion and its effects on host responses focusing on changes in the global epigenome and transcriptome. Using a multiomic approach, we analyze epigenomic alterations of H3K27ac and global transcriptomic alterations sustained within a hypoxia and normoxia conditioned CRC cell line HCT116 at 24 h following initial infection with F. nucleatum. Our findings reveal that intracellular F. nucleatum activates signaling pathways and biological processes in host cells similar to those induced upon hypoxia conditioning in the absence of infection. Furthermore, we show that a hypoxic TME favors F. nucleatum invasion and persistence and therefore infection under hypoxia may amplify malignant transformation by exacerbating the effects induced by hypoxia alone. These results motivate future studies to investigate host-microbe interactions in tumor tissue relevant conditions that more accurately define parameters for targeted cancer therapies.
Collapse
Affiliation(s)
- Barath Udayasuryan
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Zirui Zhou
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Raffae N Ahmad
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Polina Sobol
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Chengyu Deng
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Tam T D Nguyen
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
| | - Shivanie Kodikalla
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Ryan Morrison
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Ishan Goswami
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Daniel J Slade
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
| | - Scott S Verbridge
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA
| | - Chang Lu
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
16
|
Su L, Yang R, Sheng Y, Ullah S, Zhao Y, Shunjiayi H, Zhao Z, Wang Q. Insights into the oral microbiota in human systemic cancers. Front Microbiol 2024; 15:1369834. [PMID: 38756728 PMCID: PMC11098135 DOI: 10.3389/fmicb.2024.1369834] [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: 01/13/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
The oral cavity stands as one of the pivotal interfaces facilitating the intricate interaction between the human body and the external environment. The impact of diverse oral microorganisms on the emergence and progression of various systemic cancers, typified by oral cancer, has garnered increasing attention. The potential pathogenicity of oral bacteria, notably the anaerobic Porphyromonas gingivalis and Fusobacterium nucleatum, has been extensively studied and exhibits obvious correlation with different carcinoma types. Furthermore, oral fungi and viruses are closely linked to oropharyngeal carcinoma. Multiple potential mechanisms of oral microbiota-induced carcinogenesis have been investigated, including heightened inflammatory responses, suppression of the host immune system, influence on the tumor microenvironment, anti-apoptotic activity, and promotion of malignant transformation. The disturbance of microbial equilibrium and the migration of oral microbiota play a pivotal role in facilitating oncogenic functions. This review aims to comprehensively outline the pathogenic mechanisms by which oral microbiota participate in carcinogenesis. Additionally, this review delves into their potential applications in cancer prevention, screening, and treatment. It proves to be a valuable resource for researchers investigating the intricate connection between oral microbiota and systemic cancers.
Collapse
Affiliation(s)
- Lan Su
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Rui Yang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Yanan Sheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Saif Ullah
- Department of Microbiology School of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Yuheng Zhao
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Hu Shunjiayi
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuo Zhao
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Qingjing Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Conde‐Pérez K, Aja‐Macaya P, Buetas E, Trigo‐Tasende N, Nasser‐Ali M, Rumbo‐Feal S, Nión P, Arribas EM, Estévez LS, Otero‐Alén B, Noguera JF, Concha Á, Pardiñas‐López S, Carda‐Diéguez M, Gómez‐Randulfe I, Martínez‐Lago N, Ladra S, Aparicio LMA, Bou G, Mira Á, Vallejo JA, Poza M. The multispecies microbial cluster of Fusobacterium, Parvimonas, Bacteroides and Faecalibacterium as a precision biomarker for colorectal cancer diagnosis. Mol Oncol 2024; 18:1093-1122. [PMID: 38366793 PMCID: PMC11076999 DOI: 10.1002/1878-0261.13604] [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: 08/24/2023] [Revised: 12/27/2023] [Accepted: 01/26/2024] [Indexed: 02/18/2024] Open
Abstract
The incidence of colorectal cancer (CRC) has increased worldwide, and early diagnosis is crucial to reduce mortality rates. Therefore, new noninvasive biomarkers for CRC are required. Recent studies have revealed an imbalance in the oral and gut microbiomes of patients with CRC, as well as impaired gut vascular barrier function. In the present study, the microbiomes of saliva, crevicular fluid, feces, and non-neoplastic and tumor intestinal tissue samples of 93 CRC patients and 30 healthy individuals without digestive disorders (non-CRC) were analyzed by 16S rRNA metabarcoding procedures. The data revealed that Parvimonas, Fusobacterium, and Bacteroides fragilis were significantly over-represented in stool samples of CRC patients, whereas Faecalibacterium and Blautia were significantly over-abundant in the non-CRC group. Moreover, the tumor samples were enriched in well-known periodontal anaerobes, including Fusobacterium, Parvimonas, Peptostreptococcus, Porphyromonas, and Prevotella. Co-occurrence patterns of these oral microorganisms were observed in the subgingival pocket and in the tumor tissues of CRC patients, where they also correlated with other gut microbes, such as Hungatella. This study provides new evidence that oral pathobionts, normally located in subgingival pockets, can migrate to the colon and probably aggregate with aerobic bacteria, forming synergistic consortia. Furthermore, we suggest that the group composed of Fusobacterium, Parvimonas, Bacteroides, and Faecalibacterium could be used to design an excellent noninvasive fecal test for the early diagnosis of CRC. The combination of these four genera would significantly improve the reliability of a discriminatory test with respect to others that use a single species as a unique CRC biomarker.
Collapse
Affiliation(s)
- Kelly Conde‐Pérez
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Pablo Aja‐Macaya
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Elena Buetas
- Genomic and Health Department, FISABIO FoundationCenter for Advanced Research in Public HealthValenciaSpain
| | - Noelia Trigo‐Tasende
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Mohammed Nasser‐Ali
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Soraya Rumbo‐Feal
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Paula Nión
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Elsa Martín‐De Arribas
- Database Laboratory, Research Center for Information and Communication Technologies (CITIC)University of A Coruña (UDC)A CoruñaSpain
| | - Lara S. Estévez
- Pathology Service and BiobankUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Begoña Otero‐Alén
- Pathology Service and BiobankUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - José F. Noguera
- Surgery ServiceUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Ángel Concha
- Pathology Service and BiobankUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Simón Pardiñas‐López
- Periodontology and Oral Surgery, Pardiñas Medical Dental Clinic – Cell Therapy and Regenerative Medicine GroupInstitute of Biomedical Research (INIBIC)A CoruñaSpain
| | - Miguel Carda‐Diéguez
- Genomic and Health Department, FISABIO FoundationCenter for Advanced Research in Public HealthValenciaSpain
| | - Igor Gómez‐Randulfe
- Medical Oncology DepartmentUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | | | - Susana Ladra
- Database Laboratory, Research Center for Information and Communication Technologies (CITIC)University of A Coruña (UDC)A CoruñaSpain
| | - Luis M. A. Aparicio
- Medical Oncology DepartmentUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Germán Bou
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Álex Mira
- Genomic and Health Department, FISABIO FoundationCenter for Advanced Research in Public HealthValenciaSpain
| | - Juan A. Vallejo
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Margarita Poza
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
- Microbiome and Health Group, Faculty of SciencesUniversity of A Coruña (UDC)A CoruñaSpain
| |
Collapse
|
18
|
Nie F, Zhang J, Tian H, Zhao J, Gong P, Wang H, Wang S, Yang P, Yang C. The role of CXCL2-mediated crosstalk between tumor cells and macrophages in Fusobacterium nucleatum-promoted oral squamous cell carcinoma progression. Cell Death Dis 2024; 15:277. [PMID: 38637499 PMCID: PMC11026399 DOI: 10.1038/s41419-024-06640-7] [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: 11/02/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Dysbiosis of the oral microbiota is related to chronic inflammation and carcinogenesis. Fusobacterium nucleatum (Fn), a significant component of the oral microbiota, can perturb the immune system and form an inflammatory microenvironment for promoting the occurrence and progression of oral squamous cell carcinoma (OSCC). However, the underlying mechanisms remain elusive. Here, we investigated the impacts of Fn on OSCC cells and the crosstalk between OSCC cells and macrophages. 16 s rDNA sequencing and fluorescence in situ hybridization verified that Fn was notably enriched in clinical OSCC tissues compared to paracancerous tissues. The conditioned medium co-culture model validated that Fn and macrophages exhibited tumor-promoting properties by facilitating OSCC cell proliferation, migration, and invasion. Besides, Fn and OSCC cells can recruit macrophages and facilitate their M2 polarization. This crosstalk between OSCC cells and macrophages was further enhanced by Fn, thereby amplifying this positive feedback loop between them. The production of CXCL2 in response to Fn stimulation was a significant mediator. Suppression of CXCL2 in OSCC cells weakened Fn's promoting effects on OSCC cell proliferation, migration, macrophage recruitment, and M2 polarization. Conversely, knocking down CXCL2 in macrophages reversed the Fn-induced feedback effect of macrophages on the highly invasive phenotype of OSCC cells. Mechanistically, Fn activated the NF-κB pathway in both OSCC cells and macrophages, leading to the upregulation of CXCL2 expression. In addition, the SCC7 subcutaneous tumor-bearing model in C3H mice also substantiated Fn's ability to enhance tumor progression by facilitating cell proliferation, activating NF-κB signaling, up-regulating CXCL2 expression, and inducing M2 macrophage infiltration. However, these effects were reversed by the CXCL2-CXCR2 inhibitor SB225002. In summary, this study suggests that Fn contributes to OSCC progression by promoting tumor cell proliferation, macrophage recruitment, and M2 polarization. Simultaneously, the enhanced CXCL2-mediated crosstalk between OSCC cells and macrophages plays a vital role in the pro-cancer effect of Fn.
Collapse
Affiliation(s)
- Fujiao Nie
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Jie Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Haoyang Tian
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Jingjing Zhao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Pizhang Gong
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Huiru Wang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Suli Wang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Pishan Yang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China.
| | - Chengzhe Yang
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
19
|
Zepeda-Rivera M, Minot SS, Bouzek H, Wu H, Blanco-Míguez A, Manghi P, Jones DS, LaCourse KD, Wu Y, McMahon EF, Park SN, Lim YK, Kempchinsky AG, Willis AD, Cotton SL, Yost SC, Sicinska E, Kook JK, Dewhirst FE, Segata N, Bullman S, Johnston CD. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature 2024; 628:424-432. [PMID: 38509359 PMCID: PMC11006615 DOI: 10.1038/s41586-024-07182-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 02/08/2024] [Indexed: 03/22/2024]
Abstract
Fusobacterium nucleatum (Fn), a bacterium present in the human oral cavity and rarely found in the lower gastrointestinal tract of healthy individuals1, is enriched in human colorectal cancer (CRC) tumours2-5. High intratumoural Fn loads are associated with recurrence, metastases and poorer patient prognosis5-8. Here, to delineate Fn genetic factors facilitating tumour colonization, we generated closed genomes for 135 Fn strains; 80 oral strains from individuals without cancer and 55 unique cancer strains cultured from tumours from 51 patients with CRC. Pangenomic analyses identified 483 CRC-enriched genetic factors. Tumour-isolated strains predominantly belong to Fn subspecies animalis (Fna). However, genomic analyses reveal that Fna, considered a single subspecies, is instead composed of two distinct clades (Fna C1 and Fna C2). Of these, only Fna C2 dominates the CRC tumour niche. Inter-Fna analyses identified 195 Fna C2-associated genetic factors consistent with increased metabolic potential and colonization of the gastrointestinal tract. In support of this, Fna C2-treated mice had an increased number of intestinal adenomas and altered metabolites. Microbiome analysis of human tumour tissue from 116 patients with CRC demonstrated Fna C2 enrichment. Comparison of 62 paired specimens showed that only Fna C2 is tumour enriched compared to normal adjacent tissue. This was further supported by metagenomic analysis of stool samples from 627 patients with CRC and 619 healthy individuals. Collectively, our results identify the Fna clade bifurcation, show that specifically Fna C2 drives the reported Fn enrichment in human CRC and reveal the genetic underpinnings of pathoadaptation of Fna C2 to the CRC niche.
Collapse
Affiliation(s)
- Martha Zepeda-Rivera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Samuel S Minot
- Data Core, Shared Resources, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Heather Bouzek
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hanrui Wu
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Aitor Blanco-Míguez
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Dakota S Jones
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Ying Wu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elsa F McMahon
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Soon-Nang Park
- Korean Collection for Oral Microbiology and Department of Oral Biochemistry, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Yun K Lim
- Korean Collection for Oral Microbiology and Department of Oral Biochemistry, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | | | - Amy D Willis
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | | | - Ewa Sicinska
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joong-Ki Kook
- Korean Collection for Oral Microbiology and Department of Oral Biochemistry, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Floyd E Dewhirst
- Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Susan Bullman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Christopher D Johnston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
20
|
Zhu P, Meng L, Shu Y, Xu Y, Liu W, Bi Y, Xu J, Meng L, Li Y. Fabrication of hyaluronic acid-inulin coated Enterococcus faecium for colon-targeted delivery to fight Fusobacterium nucleatum. Carbohydr Polym 2024; 329:121797. [PMID: 38286561 DOI: 10.1016/j.carbpol.2024.121797] [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: 11/21/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/31/2024]
Abstract
The abundance of Fusobacterium nucleatum (F. nucleatum) is highly associated with the development and poor prognosis of colorectal cancer (CRC), which is regarded as a promising target for CRC. However, until now, the novel strategy to clear F. nucleatum in the colon and CRC has not been well proposed. Herein, a probiotic strain Enterococcus faecium (E. faecium, EF47) is verified to secrete various organic acids and bacteriocins to exert superior antimicrobial activity towards F. nucleatum. However, the oral delivery of EF47 is affected by the complex digestive tract environment, so we design the hyaluronic acid-inulin (HA-IN) coated EF47 for colon-targeted delivery to fight F. nucleatum. IN can protect EF47 from the harsh gastrointestinal tract environment and is degraded specifically in the colon, acting as prebiotics to further promote the proliferation of EF47. The exposed HA can also enhance the targeting effect to the tumor area via the interaction with the CD44 receptor on the tumor cells, which is confirmed to increase the adhesive ability in tumor tissues and inhibit the growth of F. nucleatum. Therefore, this colon-targeted delivery system provides a novel platform to realize high-activity and adhesive delivery of probiotics to assist the therapeutic efficiency of CRC.
Collapse
Affiliation(s)
- Pengrong Zhu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Lingtong Meng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Yue Shu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Yuqiao Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Weiming Liu
- Department of Research, Biosan Biotech Co., Ltd, Lishui, 323000, Zhejiang Province, China
| | - Yong Bi
- Department of Pharmaceutical Laboratory, Anhui Sunhere Pharmaceutical Excipients Co., Ltd., Huainan, 232000, Anhui Province, China
| | - Jian Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China.
| | - Lijuan Meng
- Department of Geriatric Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China; Department of Pharmaceutical Laboratory, Anhui Sunhere Pharmaceutical Excipients Co., Ltd., Huainan, 232000, Anhui Province, China; Department of Research, Biosan Biotech Co., Ltd, Lishui, 323000, Zhejiang Province, China.
| |
Collapse
|
21
|
Li L, Chandra V, McAllister F. Tumor-resident microbes: the new kids on the microenvironment block. Trends Cancer 2024; 10:347-355. [PMID: 38388213 PMCID: PMC11006566 DOI: 10.1016/j.trecan.2023.12.002] [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: 07/17/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 02/24/2024]
Abstract
Tumor-resident microbes (TRM) are an integral component of the tumor microenvironment (TME). TRM can influence tumor growth, distant dissemination, and response to therapies by interfering with molecular pathways in tumor cells as well as with other components of the TME. Novel technologies are improving the identification and visualization of cell type-specific microbes in the TME. The mechanisms that mediate the role of TRM at the primary tumors and metastatic sites are being elucidated. This knowledge is providing novel perspectives for targeting microbes or using microbial interventions for cancer interception or therapy.
Collapse
Affiliation(s)
- Le Li
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vidhi Chandra
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Swanton C, Bernard E, Abbosh C, André F, Auwerx J, Balmain A, Bar-Sagi D, Bernards R, Bullman S, DeGregori J, Elliott C, Erez A, Evan G, Febbraio MA, Hidalgo A, Jamal-Hanjani M, Joyce JA, Kaiser M, Lamia K, Locasale JW, Loi S, Malanchi I, Merad M, Musgrave K, Patel KJ, Quezada S, Wargo JA, Weeraratna A, White E, Winkler F, Wood JN, Vousden KH, Hanahan D. Embracing cancer complexity: Hallmarks of systemic disease. Cell 2024; 187:1589-1616. [PMID: 38552609 DOI: 10.1016/j.cell.2024.02.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 04/02/2024]
Abstract
The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.
Collapse
Affiliation(s)
- Charles Swanton
- The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - Elsa Bernard
- The Francis Crick Institute, London, UK; INSERM U981, Gustave Roussy, Villejuif, France
| | | | - Fabrice André
- INSERM U981, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France; Paris Saclay University, Kremlin-Bicetre, France
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Allan Balmain
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Susan Bullman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Ayelet Erez
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gerard Evan
- The Francis Crick Institute, London, UK; Kings College London, London, UK
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Andrés Hidalgo
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Area of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Johanna A Joyce
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Katja Lamia
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA; Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Department of Medical Oncology, The University of Melbourne, Parkville, VIC, Australia
| | | | - Miriam Merad
- Department of immunology and immunotherapy, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kathryn Musgrave
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK; Department of Haematology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ketan J Patel
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sergio Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Jennifer A Wargo
- Department of Surgical Oncology, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashani Weeraratna
- Sidney Kimmel Cancer Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA; Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton, NJ, USA
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuro-oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - John N Wood
- Molecular Nociception Group, WIBR, University College London, London, UK
| | | | - Douglas Hanahan
- Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Swiss institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland.
| |
Collapse
|
23
|
Mishra Y, Ranjan A, Mishra V, Chattaraj A, Aljabali AAA, El-Tanani M, Hromić-Jahjefendić A, Uversky VN, Tambuwala MM. The role of the gut microbiome in gastrointestinal cancers. Cell Signal 2024; 115:111013. [PMID: 38113978 DOI: 10.1016/j.cellsig.2023.111013] [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: 09/11/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
The gut microbiota present in the human digestive system is incredibly varied and is home to trillions of microorganisms. The gut microbiome is shaped at birth, while numerous genetic, dietary, and environmental variables primarily influence the microbiome composition. The importance of gut microbiota on host health is becoming more widely acknowledged. Digestion, intestinal permeability, and immunological and metabolism responses can all be affected by changes in the composition and function of the gut microbiota. There is mounting evidence that the microbial population's complex traits are important biomarkers and indicators of patient outcomes in cancer and its therapies. Numerous studies have demonstrated that changed commensal gut microorganisms contribute to the development and spread of cancer through various routes. Despite the ongoing controversy surrounding the gut microbiome and gastrointestinal cancer, accumulating evidence points to a potentially far more intricate connection than a simple cause-and-effect relationship. SIMPLE SUMMARY: Due to their high frequency and fatality rate, gastrointestinal cancers are regarded as a severe public health issue with complex medical and economic burdens. The gut microbiota may directly or indirectly interact with existing therapies like immunotherapy and chemotherapy, affecting how well a treatment works. The gut microbiome influences the immune response's activity, function, and development. Generally, certain gut bacteria impact the antitumor actions during cancer by creating particular metabolites or triggering T-cell responses. Yet, certain bacterial species have been found to promote cellular proliferation and metastasis in cancer, and comprehending these interactions in the context of cancer may help identify possible treatment targets. Notwithstanding the improvements in the field, additional research is still required to comprehend the underlying processes, examine the effects on existing therapies, and pinpoint certain bacteria and immune cells that can cause this interaction.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A A Aljabali
- Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Alkhama Medical and Health Sciences University, United Arab Emirates
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, England, United Kingdom.
| |
Collapse
|
24
|
Yadav D, Sainatham C, Filippov E, Kanagala SG, Ishaq SM, Jayakrishnan T. Gut Microbiome-Colorectal Cancer Relationship. Microorganisms 2024; 12:484. [PMID: 38543535 PMCID: PMC10974515 DOI: 10.3390/microorganisms12030484] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 11/12/2024] Open
Abstract
Traditionally, the role of gut dysbiosis was thought to be limited to pathologies like Clostridioides difficile infection, but studies have shown its role in other intestinal and extraintestinal pathologies. Similarly, recent studies have surfaced showing the strong potential role of the gut microbiome in colorectal cancer, which was traditionally attributed mainly to sporadic or germline mutations. Given that it is the third most common cancer and the second most common cause of cancer-related mortality, 78 grants totaling more than USD 28 million have been granted to improve colon cancer management since 2019. Concerted efforts by several of these studies have identified specific bacterial consortia inducing a proinflammatory environment and promoting genotoxin production, causing the induction or progression of colorectal cancer. In addition, changes in the gut microbiome have also been shown to alter the response to cancer chemotherapy and immunotherapy, thus changing cancer prognosis. Certain bacteria have been identified as biomarkers to predict the efficacy of antineoplastic medications. Given these discoveries, efforts have been made to alter the gut microbiome to promote a favorable diversity to improve cancer progression and the response to therapy. In this review, we expand on the gut microbiome, its association with colorectal cancer, and antineoplastic medications. We also discuss the evolving paradigm of fecal microbiota transplantation in the context of colorectal cancer management.
Collapse
Affiliation(s)
- Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, 2401 W Belvedere Ave, Baltimore, MD 21215, USA (E.F.); (S.M.I.)
| | - Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, 2401 W Belvedere Ave, Baltimore, MD 21215, USA (E.F.); (S.M.I.)
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, 2401 W Belvedere Ave, Baltimore, MD 21215, USA (E.F.); (S.M.I.)
| | - Sai Gautham Kanagala
- Department of Internal Medicine, NYC Health + Hospital/Metropolitan, New York, NY 10029, USA
| | - Syed Murtaza Ishaq
- Department of Internal Medicine, Sinai Hospital of Baltimore, 2401 W Belvedere Ave, Baltimore, MD 21215, USA (E.F.); (S.M.I.)
| | - Thejus Jayakrishnan
- Division of Hematology and Oncology, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
25
|
Yarahmadi A, Afkhami H. The role of microbiomes in gastrointestinal cancers: new insights. Front Oncol 2024; 13:1344328. [PMID: 38361500 PMCID: PMC10867565 DOI: 10.3389/fonc.2023.1344328] [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: 12/01/2023] [Accepted: 12/20/2023] [Indexed: 02/17/2024] Open
Abstract
Gastrointestinal (GI) cancers constitute more than 33% of new cancer cases worldwide and pose a considerable burden on public health. There exists a growing body of evidence that has systematically recorded an upward trajectory in GI malignancies within the last 5 to 10 years, thus presenting a formidable menace to the health of the human population. The perturbations in GI microbiota may have a noteworthy influence on the advancement of GI cancers; however, the precise mechanisms behind this association are still not comprehensively understood. Some bacteria have been observed to support cancer development, while others seem to provide a safeguard against it. Recent studies have indicated that alterations in the composition and abundance of microbiomes could be associated with the progression of various GI cancers, such as colorectal, gastric, hepatic, and esophageal cancers. Within this comprehensive analysis, we examine the significance of microbiomes, particularly those located in the intestines, in GI cancers. Furthermore, we explore the impact of microbiomes on various treatment modalities for GI cancer, including chemotherapy, immunotherapy, and radiotherapy. Additionally, we delve into the intricate mechanisms through which intestinal microbes influence the efficacy of GI cancer treatments.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
26
|
Zhou Q, Meng Q. Insights into the Microbial Composition of Intratumoral, Reproductive Tract, and Gut Microbiota in Ovarian Cancer Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1452:107-118. [PMID: 38805127 DOI: 10.1007/978-3-031-58311-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
According to the latest global cancer data, ovarian cancer is the deadliest among all gynecological malignant tumors and ranks fifth in terms of mortality. Its etiology and pathogenesis are unknown, and the 5-year survival rate of patients with advanced ovarian cancer is only 40% (Sung et al. CA Cancer J Clin 71:209-49, 2021). Recent research has shown that the human microbiota plays a crucial role in the development and progression of tumors, including ovarian cancer. Numerous studies have highlighted the complex connections between the reproductive tract microbiota, intestinal microbiota, and ovarian cancer (Jacobson et al. PeerJ 9:e11574, 2021). Therefore, this chapter will delve into composition, function, and the correlation between microbiota and immunity in the field of ovarian cancer microbiota, as well as the potential of bacteria in therapeutics and diagnostics of ovarian cancer.
Collapse
Affiliation(s)
- Qian Zhou
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China.
| | - Qingren Meng
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
27
|
Song Q, Zheng Y, Zhong G, Wang S, He C, Li M. Application of Nanoparticles in the Diagnosis and Treatment of Colorectal Cancer. Anticancer Agents Med Chem 2024; 24:1305-1326. [PMID: 39129164 PMCID: PMC11497148 DOI: 10.2174/0118715206323900240807110122] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024]
Abstract
Colorectal cancer is a common malignant tumor with high morbidity and mortality rates, imposing a huge burden on both patients and the healthcare system. Traditional treatments such as surgery, chemotherapy and radiotherapy have limitations, so finding more effective diagnostic and therapeutic tools is critical to improving the survival and quality of life of colorectal cancer patients. While current tumor targeting research mainly focuses on exploring the function and mechanism of molecular targets and screening for excellent drug targets, it is crucial to test the efficacy and mechanism of tumor cell therapy that targets these molecular targets. Selecting the appropriate drug carrier is a key step in effectively targeting tumor cells. In recent years, nanoparticles have gained significant interest as gene carriers in the field of colorectal cancer diagnosis and treatment due to their low toxicity and high protective properties. Nanoparticles, synthesized from natural or polymeric materials, are NM-sized particles that offer advantages such as low toxicity, slow release, and protection of target genes during delivery. By modifying nanoparticles, they can be targeted towards specific cells for efficient and safe targeting of tumor cells. Numerous studies have demonstrated the safety, efficiency, and specificity of nanoparticles in targeting tumor cells, making them a promising gene carrier for experimental and clinical studies. This paper aims to review the current application of nanoparticles in colorectal cancer diagnosis and treatment to provide insights for targeted therapy for colorectal cancer while also highlighting future prospects for nanoparticle development.
Collapse
Affiliation(s)
- Qiuyu Song
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifeng Zheng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
28
|
Kim JH, Yu J, Kim DK, Lee S, Lee SH, Ahn BK, Kim TI, Park SJ. Tumor microbiome analysis provides prognostic value for patients with stage III colorectal cancer. Front Oncol 2023; 13:1212812. [PMID: 37965445 PMCID: PMC10641399 DOI: 10.3389/fonc.2023.1212812] [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: 04/27/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
INTRODUCTION Although patients with colorectal cancer (CRC) can receive optimal treatment, the risk of recurrence remains. This study aimed to evaluate whether the tumor microbiome can be a predictor of recurrence in patients with stage III CRC. METHODS Using 16S rRNA gene sequencing, we analyzed the microbiomes of tumor and adjacent tissues acquired during surgery in 65 patients with stage III CRC and evaluated the correlation of the tissue microbiome with CRC recurrence. Additionally, the tumor tissue microbiome data of 71 patients with stage III CRC from another center were used as a validation set. RESULTS The microbial diversity and abundance significantly differed between tumor and adjacent tissues. In particular, Streptococcus and Gemella were more abundant in tumor tissue samples than in adjacent tissue samples. The microbial diversity and abundance in tumor and adjacent tissues did not differ according to the presence of recurrence, except for one genus in the validation set. Logistic regression analysis revealed that a recurrence prediction model including tumor tissue microbiome data had a better prediction performance than clinical factors (area under the curve [AUC] 0.846 vs. 0.679, p = 0.009), regardless of sex (male patients: AUC 0.943 vs. 0.818, p = 0.043; female patients: AUC 0.885 vs. 0.590, p = 0.017). When this prediction model was applied to the validation set, it had a higher AUC value than clinical factors in female patients. CONCLUSION Our results suggest that the tumor microbiome of patients with CRC be a potential predictor of postoperative disease recurrence.
Collapse
Affiliation(s)
- Jae Hyun Kim
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Republic of Korea
| | - Jongwook Yu
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong Keon Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seunghun Lee
- Department of Colorectal Surgery, Kosin University College of Medicine, Busan, Republic of Korea
| | - Seung Hyun Lee
- Department of Colorectal Surgery, Kosin University College of Medicine, Busan, Republic of Korea
| | - Byung Kwon Ahn
- Department of Colorectal Surgery, Kosin University College of Medicine, Busan, Republic of Korea
| | - Tae Il Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seun Ja Park
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
29
|
Schorr L, Mathies M, Elinav E, Puschhof J. Intracellular bacteria in cancer-prospects and debates. NPJ Biofilms Microbiomes 2023; 9:76. [PMID: 37813921 PMCID: PMC10562400 DOI: 10.1038/s41522-023-00446-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
Recent evidence suggests that some human cancers may harbor low-biomass microbial ecosystems, spanning bacteria, viruses, and fungi. Bacteria, the most-studied kingdom in this context, are suggested by these studies to localize within cancer cells, immune cells and other tumor microenvironment cell types, where they are postulated to impact multiple cancer-related functions. Herein, we provide an overview of intratumoral bacteria, while focusing on intracellular bacteria, their suggested molecular activities, communication networks, host invasion and evasion strategies, and long-term colonization capacity. We highlight how the integration of sequencing-based and spatial techniques may enable the recognition of bacterial tumor niches. We discuss pitfalls, debates and challenges in decisively proving the existence and function of intratumoral microbes, while reaching a mechanistic elucidation of their impacts on tumor behavior and treatment responses. Together, a causative understanding of possible roles played by intracellular bacteria in cancer may enable their future utilization in diagnosis, patient stratification, and treatment.
Collapse
Affiliation(s)
- Lena Schorr
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Marius Mathies
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany.
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Jens Puschhof
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
30
|
White MG, Damania A, Alshenaifi J, Sahasrabhojane P, Peacock O, Losh J, Wong MC, Lutter-Berkova Z, Chang GJ, Futreal A, Wargo JA, Ajami NJ, Kopetz S, You YN. Young-onset Rectal Cancer: Unique Tumoral Microbiome and Correlation With Response to Neoadjuvant Therapy. Ann Surg 2023; 278:538-548. [PMID: 37465976 PMCID: PMC10528779 DOI: 10.1097/sla.0000000000006015] [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] [Indexed: 07/20/2023]
Abstract
OBJECTIVE External exposures, the host, and the microbiome interact in oncology. We aimed to investigate tumoral microbiomes in young-onset rectal cancers (YORCs) for profiles potentially correlative with disease etiology and biology. BACKGROUND YORC is rapidly increasing, with 1 in 4 new rectal cancer cases occurring under the age of 50 years. Its etiology is unknown. METHODS YORC (<50 y old) or later-onset rectal cancer (LORC, ≥50 y old) patients underwent pretreatment biopsied of tumor and tumor-adjacent normal (TAN) tissue. After whole genome sequencing, metagenomic analysis quantified microbial communities comparing tumors versus TANs and YORCs versus LORCs, controlling for multiple testing. Response to neoadjuvant therapy (NT) was categorized as major pathological response (MPR, ≤10% residual viable tumor) versus non-MPR. RESULTS Our 107 tumors, 75 TANs from 37 (35%) YORCs, and 70 (65%) LORCs recapitulated bacterial species were previously associated with colorectal cancers (all P <0.0001). YORC and LORC tumoral microbiome signatures were distinct. After NT, 13 patients (12.4%) achieved complete pathologic response, whereas MPR occurred in 47 patients (44%). Among YORCs, MPR was associated with Fusobacterium nucleaum , Bacteroides dorei, and Ruminococcus bromii (all P <0.001), but MPR in LORC was associated with R. bromii ( P <0.001). Network analysis of non-MPR tumors demonstrated a preponderance of oral bacteria not observed in MPR tumors. CONCLUSIONS Microbial signatures were distinct between YORC and LORC. Failure to achieve an MPR was associated with oral bacteria in tumors. These findings urge further studies to decipher correlative versus mechanistic associations but suggest a potential for microbial modulation to augment current treatments.
Collapse
Affiliation(s)
- Michael G. White
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ashish Damania
- Platform for Innovative Microbiome and Translational research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jumanah Alshenaifi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pranoti Sahasrabhojane
- Platform for Innovative Microbiome and Translational research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Oliver Peacock
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jillian Losh
- Platform for Innovative Microbiome and Translational research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew C Wong
- Platform for Innovative Microbiome and Translational research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zuzana Lutter-Berkova
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George J. Chang
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A. Wargo
- Platform for Innovative Microbiome and Translational research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nadim J. Ajami
- Platform for Innovative Microbiome and Translational research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y. Nancy You
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
31
|
Cuesta-Borràs E, Salvans C, Arqués O, Chicote I, Ramírez L, Cabellos L, Martínez-Quintanilla J, Mur-Espinosa A, García-Álvarez A, Hernando J, Tejedor JR, Mirallas O, Élez E, Fraga MF, Tabernero J, Nuciforo P, Capdevila J, Palmer HG, Puig I. DPPA3-HIF1α axis controls colorectal cancer chemoresistance by imposing a slow cell-cycle phenotype. Cell Rep 2023; 42:112927. [PMID: 37537841 DOI: 10.1016/j.celrep.2023.112927] [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: 12/29/2022] [Revised: 06/22/2023] [Accepted: 07/17/2023] [Indexed: 08/05/2023] Open
Abstract
Tumor relapse is linked to rapid chemoresistance and represents a bottleneck for cancer therapy success. Engagement of a reduced proliferation state is a non-mutational mechanism exploited by cancer cells to bypass therapy-induced cell death. Through combining functional pulse-chase experiments in engineered cells and transcriptomic analyses, we identify DPPA3 as a master regulator of slow-cycling and chemoresistant phenotype in colorectal cancer (CRC). We find a vicious DPPA3-HIF1α feedback loop that downregulates FOXM1 expression via DNA methylation, thereby delaying cell-cycle progression. Moreover, downregulation of HIF1α partially restores a chemosensitive proliferative phenotype in DPPA3-overexpressing cancer cells. In cohorts of CRC patient samples, DPPA3 overexpression acts as a predictive biomarker of chemotherapeutic resistance that subsequently requires reduction in its expression to allow metastatic outgrowth. Our work demonstrates that slow-cycling cancer cells exploit a DPPA3/HIF1α axis to support tumor persistence under therapeutic stress and provides insights on the molecular regulation of disease progression.
Collapse
Affiliation(s)
- Estefania Cuesta-Borràs
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Cándida Salvans
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Oriol Arqués
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Irene Chicote
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; CIBERONC, 08029 Madrid, Spain
| | - Lorena Ramírez
- Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Laia Cabellos
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | | | - Alex Mur-Espinosa
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Alejandro García-Álvarez
- Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Jorge Hernando
- Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Juan Ramón Tejedor
- Nanomaterials and Nanotechnology Research Center (CINN), Spanish National Research Council (CSIC), Health Research Institute of the Principality of Asturias (ISPA), Spanish Biomedical Research Network in Rare Diseases (CIBERER), Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Asturias, Spain
| | - Oriol Mirallas
- Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Elena Élez
- CIBERONC, 08029 Madrid, Spain; Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center (CINN), Spanish National Research Council (CSIC), Health Research Institute of the Principality of Asturias (ISPA), Spanish Biomedical Research Network in Rare Diseases (CIBERER), Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Asturias, Spain
| | - Josep Tabernero
- CIBERONC, 08029 Madrid, Spain; Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; UVic-UCC, IOB-Quiron, 08023 Barcelona, Spain
| | - Paolo Nuciforo
- CIBERONC, 08029 Madrid, Spain; Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Jaume Capdevila
- Gastrointestinal and Endocrine Tumors Group, Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; IOB-Teknon, 08023 Barcelona, Spain
| | - Héctor G Palmer
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; CIBERONC, 08029 Madrid, Spain.
| | - Isabel Puig
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; CIBERONC, 08029 Madrid, Spain.
| |
Collapse
|
32
|
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: 10] [Impact Index Per Article: 5.0] [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
|
33
|
Queen J, Shaikh F, Sears CL. Understanding the mechanisms and translational implications of the microbiome for cancer therapy innovation. NATURE CANCER 2023; 4:1083-1094. [PMID: 37525016 DOI: 10.1038/s43018-023-00602-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
The intersection of the microbiota and cancer and the mechanisms that define these interactions are a fascinating, rapidly evolving area of cancer biology and therapeutics. Here we present recent insights into the mechanisms by which specific bacteria or their communities contribute to carcinogenesis and discuss the bidirectional interplay between microbiota and host gene or epigenome signaling. We conclude with comments on manipulation of the microbiota for the therapeutic benefit of patients with cancer.
Collapse
Affiliation(s)
- Jessica Queen
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fyza Shaikh
- Cancer Immunology Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cancer Immunology Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Microbiology and Molecular Immunology, Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
34
|
Inchingolo AM, Malcangi G, Piras F, Palmieri G, Settanni V, Riccaldo L, Morolla R, Buongiorno S, de Ruvo E, Inchingolo AD, Mancini A, Inchingolo F, Dipalma G, Benagiano S, Tartaglia GM, Patano A. Precision Medicine on the Effects of Microbiota on Head-Neck Diseases and Biomarkers Diagnosis. J Pers Med 2023; 13:933. [PMID: 37373922 DOI: 10.3390/jpm13060933] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Precision medicine using highly precise technologies and big data has produced personalised medicine with rapid and reliable diagnoses and targeted therapies. The most recent studies have directed precision medicine into the study of tumours. The application of precision medicine in the oral microbiota can be used both in the field of prevention and treatment in the strictly dental field. This article aims to evaluate the interaction between microbiota and oral cancer and the presence of biomarkers as risk predictors. MATERIALS AND METHODS A literature search of PubMed, Scopus, and Web of Science was performed analysing the various interactions between microorganisms, biomarkers, and oral cancer. RESULTS After screening processes, 21 articles were selected for qualitative analysis. CONCLUSION The correlation between oral diseases/cancers and changes in the microbiota explains the increasing utility of precision medicine in enhancing diagnosis and adapting treatment on the individual components of the microbiota. Diagnosing and treating oral diseases and cancers through precision medicine gives, as well as economic advantages to the health care system, predictable and rapid management of the patient.
Collapse
Affiliation(s)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Fabio Piras
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Giulia Palmieri
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Vito Settanni
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Lilla Riccaldo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Roberta Morolla
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Silvio Buongiorno
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Elisabetta de Ruvo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | | | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Stefania Benagiano
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20122 Milan, Italy
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Assunta Patano
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70121 Bari, Italy
| |
Collapse
|
35
|
Pani G. Fusobacterium & Co. at the Stem of Cancer: Microbe-Cancer Stem Cell Interactions in Colorectal Carcinogenesis. Cancers (Basel) 2023; 15:cancers15092583. [PMID: 37174049 PMCID: PMC10177588 DOI: 10.3390/cancers15092583] [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: 04/12/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Adult stem cells lie at the crossroads of tissue repair, inflammation, and malignancy. Intestinal microbiota and microbe-host interactions are pivotal to maintaining gut homeostasis and response to injury, and participate in colorectal carcinogenesis. Yet, limited knowledge is available on whether and how bacteria directly crosstalk with intestinal stem cells (ISC), particularly cancerous stem-like cells (CR-CSC), as engines for colorectal cancer initiation, maintenance, and metastatic dissemination. Among several bacterial species alleged to initiate or promote colorectal cancer (CRC), the pathobiont Fusobacterium Nucleatum has recently drawn significant attention for its epidemiologic association and mechanistic linkage with the disease. We will therefore focus on current evidence for an F. nucleatum-CRCSC axis in tumor development, highlighting the commonalities and differences between F. nucleatum-associated colorectal carcinogenesis and gastric cancer driven by Helicobacter Pylori. We will explore the diverse facets of the bacteria-CSC interaction, analyzing the signals and pathways whereby bacteria either confer "stemness" properties to tumor cells or primarily target stem-like elements within the heterogeneous tumor cell populations. We will also discuss the extent to which CR-CSC cells are competent for innate immune responses and participate in establishing a tumor-promoting microenvironment. Finally, by capitalizing on the expanding knowledge of how the microbiota and ISC crosstalk in intestinal homeostasis and response to injury, we will speculate on the possibility that CRC arises as an aberrant repair response promoted by pathogenic bacteria upon direct stimulation of intestinal stem cells.
Collapse
Affiliation(s)
- Giovambattista Pani
- Department of Translational Medicine and Surgery, Section of General Pathology, Faculty of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, L. go A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
36
|
Palmieri O, Castellana S, Latiano A, Latiano T, Gentile A, Panza A, Nardella M, Ciardiello D, Latiano TP, Corritore G, Mazza T, Perri F, Biscaglia G. Mucosal Microbiota from Colorectal Cancer, Adenoma and Normal Epithelium Reveals the Imprint of Fusobacterium nucleatum in Cancerogenesis. Microorganisms 2023; 11:1147. [PMID: 37317121 DOI: 10.3390/microorganisms11051147] [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: 03/15/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 06/16/2023] Open
Abstract
An increasing amount of evidence suggests the emerging role of the gut microbiota in the development of colorectal cancer (CRC). This study aimed to elucidate the architecture of microbial communities within normal and neoplastic colonic mucosa. METHODS Microbiota were analyzed by NGS and by an ensemble of metagenomics analysis tools in a total of 69 tissues from 9 patients with synchronous colorectal neoplasia and adenomas (27 specimens: 9 from normal tissues, 9 adenomas, and 9 tumours), 16 patients with only colonic adenomas (32 specimens: 16 from normal tissues and 16 adenomas), and from healthy subjects (10 specimens of normal mucosa). RESULTS Weak differences were observed in alpha and beta metrics among the synchronous tissues from CRC and controls. Through pairwise differential abundance analyses of sample groups, an increasing trend of Rikenellaceae, Pseudomonas and Fusobacterium, and decreasing trends of Staphylococcus, Actinobacillus and Gemmiger were observed in CRC, while Staphylococcus and Bifidobacterium were decreased in patients with only adenomas. At RT-qPCR analysis, Fusobacterium nucleatum was significantly enriched in all the tissues of subjects with synchronous colorectal neoplasia. CONCLUSION Our findings provide a comprehensive view of the human mucosa-associated gut microbiota, emphasizing global microbial diversity mostly in synchronous lesions and proving the constant presence of Fusobacterium nucleatum, with its ability to drive carcinogenesis.
Collapse
Affiliation(s)
- Orazio Palmieri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Stefano Castellana
- Unit of Bioinformatics, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Anna Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Tiziana Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Annamaria Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Anna Panza
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Marianna Nardella
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Davide Ciardiello
- Division of Oncology, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
- Division of Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Tiziana Pia Latiano
- Division of Oncology, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Giuseppe Corritore
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Tommaso Mazza
- Unit of Bioinformatics, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Francesco Perri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Giuseppe Biscaglia
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
37
|
Jiang SS, Xie YL, Xiao XY, Kang ZR, Lin XL, Zhang L, Li CS, Qian Y, Xu PP, Leng XX, Wang LW, Tu SP, Zhong M, Zhao G, Chen JX, Wang Z, Liu Q, Hong J, Chen HY, Chen YX, Fang JY. Fusobacterium nucleatum-derived succinic acid induces tumor resistance to immunotherapy in colorectal cancer. Cell Host Microbe 2023; 31:781-797.e9. [PMID: 37130518 DOI: 10.1016/j.chom.2023.04.010] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/05/2022] [Accepted: 04/06/2023] [Indexed: 05/04/2023]
Abstract
Immune checkpoint blockade therapy with anti-PD-1 monoclonal antibody (mAb) is a treatment for colorectal cancer (CRC). However, some patients remain unresponsive to PD-1 blockade. The gut microbiota has been linked to immunotherapy resistance through unclear mechanisms. We found that patients with metastatic CRC who fail to respond to immunotherapy had a greater abundance of Fusobacterium nucleatum and increased succinic acid. Fecal microbiota transfer from responders with low F. nucleatum, but not F. nucleatum-high non-responders, conferred sensitivity to anti-PD-1 mAb in mice. Mechanistically, F. nucleatum-derived succinic acid suppressed the cGAS-interferon-β pathway, consequently dampening the antitumor response by limiting CD8+ T cell trafficking to the tumor microenvironment (TME) in vivo. Treatment with the antibiotic metronidazole reduced intestinal F. nucleatum abundance, thereby decreasing serum succinic acid levels and resensitizing tumors to immunotherapy in vivo. These findings indicate that F. nucleatum and succinic acid induce tumor resistance to immunotherapy, offering insights into microbiota-metabolite-immune crosstalk in CRC.
Collapse
Affiliation(s)
- Shan-Shan Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Yi-Le Xie
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiu-Ying Xiao
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zi-Ran Kang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiao-Lin Lin
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Zhang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Chu-Shu Li
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Yun Qian
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Ping-Ping Xu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiao-Xu Leng
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Li-Wei Wang
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shui-Ping Tu
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jin-Xian Chen
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jie Hong
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Hao-Yan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| |
Collapse
|
38
|
Crowder SL, Jim HSL, Hogue S, Carson TL, Byrd DA. Gut microbiome and cancer implications: Potential opportunities for fermented foods. Biochim Biophys Acta Rev Cancer 2023; 1878:188897. [PMID: 37086870 DOI: 10.1016/j.bbcan.2023.188897] [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: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
There is a critical opportunity to improve response to immunotherapies and overall cancer survivorship via dietary interventions targeted to modify the gut microbiome, and in turn, potentially enhance anti-cancer immunity. A promising dietary intervention is fermented foods, which may alter gut microbiome composition and, in turn, improve immunity. In this article, we summarize the state of the literature pertaining to the gut microbiome and response to immunotherapy and other cancer treatments, potential clinical implications of utilizing a fermented foods dietary approach to improve cancer treatment outcomes, and existing gaps in the literature regarding the implementation of fermented food interventions among individuals with cancer or with a history of cancer. This review synthesizes a compelling rationale across different disciplines to lay a roadmap for future fermented food dietary intervention research aimed at modulating the gut microbiome to reduce cancer burden.
Collapse
Affiliation(s)
- Sylvia L Crowder
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Heather S L Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Stephanie Hogue
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Tiffany L Carson
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Doratha A Byrd
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
39
|
Little A, Tangney M, Tunney MM, Buckley NE. Fusobacterium nucleatum: a novel immune modulator in breast cancer? Expert Rev Mol Med 2023; 25:e15. [PMID: 37009688 PMCID: PMC10407221 DOI: 10.1017/erm.2023.9] [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: 10/28/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 04/04/2023]
Abstract
Breast cancer was the most commonly diagnosed cancer worldwide in 2020. Greater understanding of the factors which promote tumour progression, metastatic development and therapeutic resistance is needed. In recent years, a distinct microbiome has been detected in the breast, a site previously thought to be sterile. Here, we review the clinical and molecular relevance of the oral anaerobic bacterium Fusobacterium nucleatum in breast cancer. F. nucleatum is enriched in breast tumour tissue compared with matched healthy tissue and has been shown to promote mammary tumour growth and metastatic progression in mouse models. Current literature suggests that F. nucleatum modulates immune escape and inflammation within the tissue microenvironment, two well-defined hallmarks of cancer. Furthermore, the microbiome, and F. nucleatum specifically, has been shown to affect patient response to therapy including immune checkpoint inhibitors. These findings highlight areas of future research needed to better understand the influence of F. nucleatum in the development and treatment of breast cancer.
Collapse
Affiliation(s)
- Alexa Little
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Mark Tangney
- Cancer Research, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Michael M. Tunney
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Niamh E. Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| |
Collapse
|
40
|
Kosumi K, Baba Y, Yamamura K, Nomoto D, Okadome K, Yagi T, Toihata T, Kiyozumi Y, Harada K, Eto K, Sawayama H, Ishimoto T, Iwatsuki M, Iwagami S, Miyamoto Y, Yoshida N, Watanabe M, Baba H. Intratumour Fusobacterium nucleatum and immune response to oesophageal cancer. Br J Cancer 2023; 128:1155-1165. [PMID: 36599917 PMCID: PMC10006219 DOI: 10.1038/s41416-022-02112-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Experimental evidence suggests a role of intratumour Fusobacterium nucleatum in the aggressive behaviour of gastrointestinal cancer through downregulating anti-tumour immunity. We investigated the relationship between intratumour F. nucleatum and immune response to oesophageal cancer. METHODS Utilising an unbiased database of 300 resected oesophageal cancers, we measured F. nucleatum DNA in tumour tissue using a quantitative polymerase chain reaction assay, and evaluated the relationship between the abundance of F. nucleatum and the densities of T cells (CD8 + , FOXP3 + and PDCD1 + ), as well as lymphocytic reaction patterns (follicle lymphocytic reaction, peritumoural lymphocytic reaction, stromal lymphocytic reaction and tumour-infiltrating lymphocytes) in oesophageal carcinoma tissue. RESULTS F. nucleatum was significantly and inversely associated only with the peritumoural lymphocytic reaction (P = 0.0002). Compared with the F. nucleatum-absent group, the F. nucleatum-high group showed a much lower level of the peritumoural lymphocytic reaction (univariable odds ratio, 0.33; 95% confidence interval, 0.16-0.65; P = 0.0004). A multivariable model yielded a similar finding (multivariable odds ratio, 0.34; 95% confidence interval 0.16-0.69; P = 0.002). CONCLUSIONS Intratumour F. nucleatum is associated with a diminished peritumoural lymphocytic reaction, providing a platform for further investigations on the potential interactive roles between intratumour F. nucleatum and host immunity.
Collapse
Affiliation(s)
- Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
- Department of Surgery, Amakusa Medical Center, 854-1 Jikiba, Kameba-machi, Amakusa, 863-0046, Japan
- Department of Next-Generation Surgical Therapy Development, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
- Department of Next-Generation Surgical Therapy Development, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kensuke Yamamura
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Daichi Nomoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kazuo Okadome
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Ave., EBRC Room 404A, Boston, MA, 02115, USA
| | - Taisuke Yagi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Tasuku Toihata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yuki Kiyozumi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hiroshi Sawayama
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
41
|
Yang S, Hao S, Ye H, Zhang X. Global research on the crosstalk between intestinal microbiome and colorectal cancer: A visualization analysis. Front Cell Infect Microbiol 2023; 13:1083987. [PMID: 37009513 PMCID: PMC10050574 DOI: 10.3389/fcimb.2023.1083987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundIncreasing evidence has shown that the intestinal microbiome (IM) is highly linked to colorectal cancer (CRC). To investigate scientific output, identify highly cited papers, and explore research hotspots and trends in the field of IM/CRC, we conducted a bibliometric and visualized analysis.MethodsA bibliographic search regarding IM/CRC research (2012-2021) was implemented on October 17, 2022. The terms attached to IM and CRC were searched for in the titles (TI), abstracts (AB), and author keywords (AK). The main information was extracted from the Web of Science Core Collection (WoSCC). Biblioshiny from R packages and VOSviewer were used for data visualization.ResultsA total of 1725 papers related to IM/CRC were retrieved. Publications on IM/CRC have grown rapidly from 2012 to 2021. China and the United States were in the leading position for publications in this field and made the most significant contributions to IM/CRC research. Shanghai Jiao Tong University and Harvard University were the most productive institutions. The high-yield authors were Yu Jun and Fang Jing Yuan. The International Journal of Molecular Sciences published the most papers, whereas Gut had the most citations. Historical citation analysis showed the evolution of IM/CRC research. Current status and hotspots were highlighted using keyword cluster analysis. The hot topics include the effect of IM on tumorigenesis, the effect of IM on CRC treatment, the role of IM in CRC screening, the mechanisms of IM involvement in CRC, and IM modulation for CRC management. Some topics, such as chemotherapy, immunotherapy, Fusobacterium nucleatum and short-chain fatty acids could be the focus of IM/CRC research in the coming years.ConclusionThis research evaluated the global scientific output of IM/CRC research and its quantitative features, identified some significant papers, and gathered information on the status and trends of IM/CRC research, which may shape future paths for academics and practitioners.
Collapse
Affiliation(s)
- Shanshan Yang
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shaodong Hao
- Spleen-Stomach Department, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Ye
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital, Beijing, China
- *Correspondence: Xuezhi Zhang, ; Hui Ye,
| | - Xuezhi Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital, Beijing, China
- *Correspondence: Xuezhi Zhang, ; Hui Ye,
| |
Collapse
|
42
|
Abstract
The microbiome (bacteria, viruses, and fungi) that exist within a patient's gastrointestinal tract and throughout their body have been increasingly understood to play a critical role in a variety of disease, including a number of cancer histologies. These microbial colonies are reflective of a patient's overall health state, their exposome, and germline genetics. In the case of colorectal adenocarcinoma, significant progress has been made in understanding the mechanism the microbiome plays beyond mere associations in both disease initiation and progression. Importantly, this improved understanding holds the potential to further identify the role these microbes play in colorectal cancer. We hope this improved understanding will be able to be leveraged in the future through either biomarkers or next-generation therapeutics to augment contemporary treatment algorithms through the manipulation of a patient's microbiome-whether through diet, antibiotics, prebiotics, or novel therapeutics. Here we review the role of the microbiome in the setting of patients with stage IV colorectal adenocarcinoma in both the development and progression or disease as well as response to therapeutics.
Collapse
Affiliation(s)
- Samuel Cass
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael G. White
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
43
|
Wang N, Fang JY. Fusobacterium nucleatum, a key pathogenic factor and microbial biomarker for colorectal cancer. Trends Microbiol 2023; 31:159-172. [PMID: 36058786 DOI: 10.1016/j.tim.2022.08.010] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/27/2023]
Abstract
Colorectal cancer (CRC), one of the most prevalent cancers, has complex etiology. The dysbiosis of intestinal bacteria has been highlighted as an important contributor to CRC. Fusobacterium nucleatum, an oral anaerobic opportunistic pathogen, is enriched in both stools and tumor tissues of patients with CRC. Therefore, F. nucleatum is considered to be a risk factor for CRC. This review summarizes the biological characteristics and the mechanisms underlying the regulatory behavior of F. nucleatum in the tumorigenesis and progression of CRC. F. nucleatum as a marker for the early warning and prognostic prediction of CRC, and as a target for prevention and treatment, is also described.
Collapse
Affiliation(s)
- Ni Wang
- Division of Gastroenterology and Hepatology, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China; NHC Key Laboratory of Digestive Diseases, Shanghai Jiao Tong University, Shanghai, China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China; NHC Key Laboratory of Digestive Diseases, Shanghai Jiao Tong University, Shanghai, China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
44
|
Bukavina L, Ginwala R, Sindhani M, Prunty M, Geynisman D, Pooja G, Valentine H, Calaway A, Brown JR, Correa A, Mishra K, Pominville R, Plimack E, Kutikov A, Ghannoum M, ElShaer M, Retuerto M, Uzzo R, Ponsky L, Abbosh PH. Role of Gut Microbiome in Neoadjuvant Chemotherapy Response in Urothelial Carcinoma: A Multi-Institutional Prospective Cohort Evaluation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.21.525021. [PMID: 36747848 PMCID: PMC9900756 DOI: 10.1101/2023.01.21.525021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Treatment with neoadjuvant chemotherapy (NAC) in muscle invasive bladder cancer (MIBC) is associated with clinical benefit in urothelial carcinoma. While extensive research evaluating role of tumor mutational expression profiles and clinicopathologic factors into chemoresponse has been published, the role of gut microbiome (GM) in bladder cancer in chemoresponse has not been thoroughly evaluated. A working knowledge of the microbiome and its effect on all forms of cancer therapy in BC is critical. Here we examine gut microbiome of bladder cancer patients undergoing NAC. Overall, there was no significant difference in alpha and beta diversity by responder status. However, analysis of fecal microbiome samples showed that a higher abundance of Bacteroides within both institutional cohorts during NAC was associated with residual disease at the time of radical cystectomy regardless of chemotherapy regimen. Group community analysis revealed presence of favorable microbial subtypes in complete responders. Finally, fecal microbial composition outperformed clinical variables in prediction of complete response (AUC 0.88 vs AUC 0.50), however, no single microbial species could be regarded as a fully consistent biomarker. Microbiome-based community signature as compared to single microbial species is more likely to be associated as the link between bacterial composition and NAC response.
Collapse
|
45
|
Holt RA. Oncomicrobial vaccines: The potential for a Fusobacterium nucleatum vaccine to improve colorectal cancer outcomes. Cell Host Microbe 2023; 31:141-145. [PMID: 36634619 DOI: 10.1016/j.chom.2022.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
There is increasing awareness of the many different ways host-microbe interactions relate to cancer initiation and progression. Vaccines designed to drive immune responses against key tumor-promoting mechanisms of oncomicrobes like F. nucleatum may provide novel and effective interventions against colorectal cancer and other diseases.
Collapse
Affiliation(s)
- Robert A Holt
- British Columbia Cancer Research Institute, Vancouver, BC, Canada; Vancouver Coastal Health Research Institute, Vancouver, BC, Canada; University of British Columbia, Vancouver, BC, Canada; Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
46
|
Dougherty MW, Jobin C. Intestinal bacteria and colorectal cancer: etiology and treatment. Gut Microbes 2023; 15:2185028. [PMID: 36927206 PMCID: PMC10026918 DOI: 10.1080/19490976.2023.2185028] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/17/2023] [Indexed: 03/18/2023] Open
Abstract
The etiology of colorectal cancer (CRC) is influenced by bacterial communities that colonize the gastrointestinal tract. These microorganisms derive essential nutrients from indigestible dietary or host-derived compounds and activate molecular signaling pathways necessary for normal tissue and immune function. Associative and mechanistic studies have identified bacterial species whose presence may increase CRC risk, including notable examples such as Fusobacterium nucleatum, Enterotoxigenic Bacteroides fragilis, and pks+ E. coli. In recent years this work has expanded in scope to include aspects of host mutational status, intra-tumoral microbial heterogeneity, transient infection, and the cumulative influence of multiple carcinogenic bacteria after sequential or co-colonization. In this review, we will provide an updated overview of how host-bacteria interactions influence CRC development, how this knowledge may be utilized to diagnose or prevent CRC, and how the gut microbiome influences CRC treatment efficacy.
Collapse
Affiliation(s)
- Michael W. Dougherty
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christian Jobin
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Infectious Diseases and Immunology, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
47
|
The conundrum of breast cancer and microbiome - A comprehensive review of the current evidence. Cancer Treat Rev 2022; 111:102470. [DOI: 10.1016/j.ctrv.2022.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/15/2022]
|
48
|
Ramai D, Salati M, Pomati G, Amoroso C, Facciorusso A, Botticelli A, Ghidini M. Antibiotics, the microbiome and gastrointestinal cancers: A causal interference? Curr Opin Pharmacol 2022; 67:102315. [PMID: 36351361 DOI: 10.1016/j.coph.2022.102315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/07/2022]
Abstract
Our understanding of the gut microbiota has significantly evolved over the last two decades. Advances in the analysis of the gut microbiome continues to reveal complex microbial communities and discoveries about their role in health and diseases, including cancer development, are continuously growing. In addition, research has demonstrated that the use of antibiotics can modulate the gut microbiota composition negatively and influence cancer treatment outcomes, suggesting that antibiotics should be avoided if possible. In this article, we review the role of the gut microbiota in the formation of GI cancers. We show that specific bacterial populations can positively or negatively affect cancer formation with specific attention given to gastric and colorectal cancer. We also review the role of microbial-targeted therapies on cancer treatment outcomes.
Collapse
Affiliation(s)
- Daryl Ramai
- Division of Gastroenterology and Hepatology, University of Utah Health, Salt Lake City, UT, USA
| | - Massimiliano Salati
- Department of Oncology and Hematology, Division of Oncology, University Hospital of Modena, Modena, Italy
| | - Giulia Pomati
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Facciorusso
- Gastroenterology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Andrea Botticelli
- Department of Radiological, Oncological, Pathological Department, La Sapienza, University of Rome, Policlinico Umberto I, Rome, Italy
| | - Michele Ghidini
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
49
|
Mei S, Deng Z, Chen Y, Ning D, Guo Y, Fan X, Wang R, Meng Y, Zhou Q, Tian X. Dysbiosis: The first hit for digestive system cancer. Front Physiol 2022; 13:1040991. [PMID: 36483296 PMCID: PMC9723259 DOI: 10.3389/fphys.2022.1040991] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/01/2022] [Indexed: 03/01/2025] Open
Abstract
Gastrointestinal cancer may be associated with dysbiosis, which is characterized by an alteration of the gut microbiota. Understanding the role of gut microbiota in the development of gastrointestinal cancer is useful for cancer prevention and gut microbiota-based therapy. However, the potential role of dysbiosis in the onset of tumorigenesis is not fully understood. While accumulating evidence has demonstrated the presence of dysbiosis in the intestinal microbiota of both healthy individuals and patients with various digestive system diseases, severe dysbiosis is often present in patients with digestive system cancer. Importantly, specific bacteria have been isolated from the fecal samples of these patients. Thus, the association between dysbiosis and the development of digestive system cancer cannot be ignored. A new model describing this relationship must be established. In this review, we postulate that dysbiosis serves as the first hit for the development of digestive system cancer. Dysbiosis-induced alterations, including inflammation, aberrant immune response, bacteria-produced genotoxins, and cellular stress response associated with genetic, epigenetic, and/or neoplastic changes, are second hits that speed carcinogenesis. This review explains the mechanisms for these four pathways and discusses gut microbiota-based therapies. The content included in this review will shed light on gut microbiota-based strategies for cancer prevention and therapy.
Collapse
Affiliation(s)
- Si Mei
- Department of Physiology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhe Deng
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yating Chen
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Dimin Ning
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yinmei Guo
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xingxing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Ruoyu Wang
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Liver Diseases, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuelin Meng
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qing Zhou
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuefei Tian
- Hunan Key Laboratory of Translational Research in Formulas and Zheng of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
50
|
LaCourse KD, Zepeda-Rivera M, Kempchinsky AG, Baryiames A, Minot SS, Johnston CD, Bullman S. The cancer chemotherapeutic 5-fluorouracil is a potent Fusobacterium nucleatum inhibitor and its activity is modified by intratumoral microbiota. Cell Rep 2022; 41:111625. [PMID: 36384132 PMCID: PMC10790632 DOI: 10.1016/j.celrep.2022.111625] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/26/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Fusobacterium nucleatum (Fn) is a dominant bacterial species in colorectal cancer (CRC) tissue that is associated with cancer progression and poorer patient prognosis. Following a small-molecule inhibitor screen of 1,846 bioactive compounds against a Fn CRC isolate, we find that 15% of inhibitors are antineoplastic agents including fluoropyrimidines. Validation of these findings reveals that 5-fluorouracil (5-FU), a first-line CRC chemotherapeutic, is a potent inhibitor of Fn CRC isolates. We also identify members of the intratumoral microbiota, including Escherichia coli, that are resistant to 5-FU. Further, CRC E. coli isolates can modify 5-FU and relieve 5-FU toxicity toward otherwise-sensitive Fn and human CRC epithelial cells. Lastly, we demonstrate that ex vivo patient CRC tumor microbiota undergo community disruption after 5-FU exposure and have the potential to deplete 5-FU levels, reducing local drug efficacy. Together, these observations argue for further investigation into the role of the CRC intratumoral microbiota in patient response to chemotherapy.
Collapse
Affiliation(s)
- Kaitlyn D LaCourse
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Martha Zepeda-Rivera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Andrew G Kempchinsky
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Alexander Baryiames
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Samuel S Minot
- Data Core, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Christopher D Johnston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA.
| | - Susan Bullman
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA.
| |
Collapse
|