1
|
Lin A, Jiang A, Huang L, Li Y, Zhang C, Zhu L, Mou W, Liu Z, Zhang J, Cheng Q, Wei T, Luo P. From chaos to order: optimizing fecal microbiota transplantation for enhanced immune checkpoint inhibitors efficacy. Gut Microbes 2025; 17:2452277. [PMID: 39826104 DOI: 10.1080/19490976.2025.2452277] [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: 10/16/2024] [Revised: 11/22/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
The integration of fecal microbiota transplantation (FMT) with immune checkpoint inhibitors (ICIs) presents a promising approach for enhancing cancer treatment efficacy and overcoming therapeutic resistance. This review critically examines the controversial effects of FMT on ICIs outcomes and elucidates the underlying mechanisms. We investigate how FMT modulates gut microbiota composition, microbial metabolite profiles, and the tumor microenvironment, thereby influencing ICIs effectiveness. Key factors influencing FMT efficacy, including donor selection criteria, recipient characteristics, and administration protocols, are comprehensively discussed. The review delineates strategies for optimizing FMT formulations and systematically monitoring post-transplant microbiome dynamics. Through a comprehensive synthesis of evidence from clinical trials and preclinical studies, we elucidate the potential benefits and challenges of combining FMT with ICIs across diverse cancer types. While some studies report improved outcomes, others indicate no benefit or potential adverse effects, emphasizing the complexity of host-microbiome interactions in cancer immunotherapy. We outline critical research directions, encompassing the need for large-scale, multi-center randomized controlled trials, in-depth microbial ecology studies, and the integration of multi-omics approaches with artificial intelligence. Regulatory and ethical challenges are critically addressed, underscoring the imperative for standardized protocols and rigorous long-term safety assessments. This comprehensive review seeks to guide future research endeavors and clinical applications of FMT-ICIs combination therapy, with the potential to improve cancer patient outcomes while ensuring both safety and efficacy. As this rapidly evolving field advances, maintaining a judicious balance between openness to innovation and cautious scrutiny is crucial for realizing the full potential of microbiome modulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lihaoyun Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Yu Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Chunyanx Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
2
|
Birebent R, Drubay D, Alves Costa Silva C, Marmorino F, Vitali G, Piccinno G, Hurtado Y, Bonato A, Belluomini L, Messaoudene M, Routy B, Fidelle M, Zalcman G, Mazieres J, Audigier-Valette C, Moro-Sibilot D, Goldwasser F, Scherpereel A, Pegliasco H, Ghiringhelli F, Reni A, Barlesi F, Albiges L, Planchard D, Martinez S, Besse B, Segata N, Cremolini C, Zitvogel L, Iebba V, Derosa L. Surrogate markers of intestinal dysfunction associated with survival in advanced cancers. Oncoimmunology 2025; 14:2484880. [PMID: 40189749 PMCID: PMC11980478 DOI: 10.1080/2162402x.2025.2484880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 02/19/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Deviations in the diversity and composition of the gut microbiota are called "gut dysbiosis". They have been linked to various chronic diseases including cancers and resistance to immunotherapy. Stool shotgun based-metagenomics informs on the ecological composition of the gut microbiota and the prevalence of homeostatic bacteria such as Akkermansia muciniphila (Akk), while determination of the serum addressin MAdCAM-1 instructs on endothelial gut barrier dysfunction. Here we examined patient survival during chemo-immuno-therapy in 955 cancer patients across four independent cohorts of non-small cell lung (NSCLC), genitourinary (GU) and colorectal (CRC) cancers, according to hallmarks of gut dysbiosis. We show that Akk prevalence represents a stable and favorable phenotype in NSCLC and CRC cancer patients. Over-dominance of Akk above the healthy threshold was observed in dismal prognosis in NSCLC and GU and mirrored an immunosuppressive gut ecosystem and excessive intestinal epithelial exfoliation in NSCLC. In CRC, the combination of a lack of Akk and low sMAdCAM-1 levels identified a subset comprising 28% of patients with reduced survival, independent of the immunoscore. We conclude that gut dysbiosis hallmarks deserve integration within the diagnosis toolbox in oncological practice.
Collapse
Affiliation(s)
- Roxanne Birebent
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Damien Drubay
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Office of Biostatistics and Epidemiology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Inserm, Université Paris-Saclay, CESP U1018, Oncostat, Villejuif, France
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Federica Marmorino
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giacomo Vitali
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- MetaGenoPolis, INRAe, Université Paris-Saclay, Jouy en Josas, France
| | | | - Yoan Hurtado
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Adele Bonato
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy
| | - Lorenzo Belluomini
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Meriem Messaoudene
- Centre Hospitalier de l’Université de Montréal (CHUM), Hematology-Oncology Division, Department of Medicine, Montréal, QC, Canada
- Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada
| | - Bertrand Routy
- Centre Hospitalier de l’Université de Montréal (CHUM), Hematology-Oncology Division, Department of Medicine, Montréal, QC, Canada
- Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada
| | - Marine Fidelle
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Gerard Zalcman
- Thoracic Oncology Department-CIC1425/CLIP2 Paris-Nord, Bichat-Claude Bernard Hospital, AP-HP, Université Paris Cité, Paris, France
| | - Julien Mazieres
- Service de Pneumologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Denis Moro-Sibilot
- Department of Thoracic Oncology, Centre Hospitalier Universitaire, Grenoble, France
| | - François Goldwasser
- INSERM U1016-CNRS UMR8104-Cochin Institute, Université Paris-Cité, Paris,France
- Department of Medical Oncology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Arnaud Scherpereel
- Department of Pulmonary and Thoracic Oncology, University of Lille, University Hospital (CHU), INSERM unit OncoThAI, Lille, France
| | | | - François Ghiringhelli
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
- Centre de Recherche INSERM LNC-UMR1241-CTM (Center of Translational and Molecular Medicine), Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Anna Reni
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Fabrice Barlesi
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Department of Clinical Oncology, Gustave Roussy, Villejuif, France
| | - Laurence Albiges
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Department of Clinical Oncology, Gustave Roussy, Villejuif, France
| | - David Planchard
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Department of Clinical Oncology, Gustave Roussy, Villejuif, France
| | - Stéphanie Martinez
- Service des Maladies Respiratoires, Centre Hospitalier d’Aix-en-Provence, Aix-en-Provence, France
| | - Benjamin Besse
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Department of Clinical Oncology, Gustave Roussy, Villejuif, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| | - Valerio Iebba
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Department of Clinical Oncology, Gustave Roussy, Villejuif, France
| | - Lisa Derosa
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France
- Faculté de Medicine, Université Paris-Saclay, Ile-de-France, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Department of Clinical Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
3
|
Talebi F, Gregucci F, Ahmed J, Ben Chetrit N, D. Brown B, Chan TA, Chand D, Constanzo J, Demaria S, I. Gabrilovich D, Golden E, Godkin A, Guha C, P. Gupta G, Hasan A, G. Herrera F, Kaufman H, Li D, A. Melcher A, McDonald S, Merghoub T, Monjazeb AM, Paris S, Pitroda S, Sadanandam A, Schaue D, Santambrogio L, Szapary P, Sage J, W. Welsh J, Wilkins A, H. Young K, Wennerberg E, Zitvogel L, Galluzzi L, Deutsch E, C. Formenti S. Updates on radiotherapy-immunotherapy combinations: Proceedings of 8th Annual ImmunoRad Conference. Oncoimmunology 2025; 14:2507856. [PMID: 40401900 PMCID: PMC12101595 DOI: 10.1080/2162402x.2025.2507856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
The annual ImmunoRad Conference has established itself as a recurrent occasion to explore the possibility of combining radiation therapy (RT) and immunotherapy (IT) for clinical cancer management. Bringing together a number of preclinical and clinical leaders in the fields of radiation oncology, immuno-oncology and IT, this annual event fosters indeed essential conversations and fruitful exchanges on how to address existing challenges to expand the therapeutic value of RT-IT combinations. The 8th edition of the ImmunoRad Conference, which has been held in October 2024 at the Weill Cornell Medical College of New York City, highlighted exciting preclinical and clinical advances at the interface between RT and IT, setting the stage for extra progress toward extended benefits for patients with an increasing variety of tumor types. Here, we critically summarize the lines of investigation that have been discussed at the occasion of the 8th Annual ImmunoRad Conference.
Collapse
Affiliation(s)
- Fereshteh Talebi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Jalal Ahmed
- Icahn Genomics Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nir Ben Chetrit
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Brian D. Brown
- Icahn Genomics Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy A. Chan
- Department of Cancer Sciences, Global Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Case Western University School of Medicine, Cleveland, OH, USA
| | | | - Julie Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Andrew Godkin
- Division of Infection and Immunity/Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Chandan Guha
- Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - Gaorav P. Gupta
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Fernanda G. Herrera
- AGORA Cancer Research Center, Swiss Cancer Center Leman, Lausanne, Switzerland
- Services of Radiation Oncology and Immuno-Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Department of Oncology, Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Donna Li
- University of Wisconsin, Madison, WI, USA
| | - Alan A. Melcher
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Sierra McDonald
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center and Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California, San Diego, CA, USA
| | | | - Sean Pitroda
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Julien Sage
- Departments of Genetics and Pediatrics, Stanford University, Stanford, California
| | - James W. Welsh
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Anna Wilkins
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Kristina H. Young
- Division of Radiation Oncology, The Oregon Clinic, Portland, OR, USA
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Eric Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Laurence Zitvogel
- Gustave Roussy, INSERM U1015, Division of Medicine, Paris-Saclay University, Center of Clinical Investigations BIOTHERIS, Villejuif, France
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, INSERM U1030, Division of Medicine, Paris-Saclay University, RHU LySAIRI “Lymphocyte-Sparing Artificial Intelligence-guided Radio-Immunotherapy”, Villejuif, France
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
4
|
Yao J, Ning B, Ding J. The gut microbiota: an emerging modulator of drug resistance in hepatocellular carcinoma. Gut Microbes 2025; 17:2473504. [PMID: 40042184 PMCID: PMC11901387 DOI: 10.1080/19490976.2025.2473504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/08/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Liver cancer is usually diagnosed at an advanced stage and is the third most common cause of cancer-related death worldwide. In addition to the lack of effective treatment options, resistance to therapeutic drugs is a major clinical challenge. The gut microbiota has recently been recognized as one of the key factors regulating host health. The microbiota and its metabolites can directly or indirectly regulate gene expression in the liver, leading to gut-liver axis dysregulation, which is closely related to liver cancer occurrence and the treatment response. Gut microbiota disturbance may participate in tumor progression and drug resistance through metabolite production, gene transfer, immune regulation, and other mechanisms. However, systematic reviews on the role of the gut microbiota in drug resistance in liver cancer are lacking. Herein, we review the relationships between the gut microbiota and the occurrence and drug resistance of hepatocellular carcinoma, summarize the emerging mechanisms underlying gut microbiota-mediated drug resistance, and propose new personalized treatment options to overcome this resistance.
Collapse
Affiliation(s)
- Jiali Yao
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Beifang Ning
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
5
|
Jia Z, Zhang Y, Cao L, Wang J, Liang H. Research hotspots and trends of immunotherapy and melanoma: A bibliometric analysis during 2014-2024. Hum Vaccin Immunother 2025; 21:2464379. [PMID: 40012099 PMCID: PMC11869780 DOI: 10.1080/21645515.2025.2464379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Over the last decade, the increasing global prevalence of melanoma has sparked growing interest in immunotherapies, which show significant potential against this form of skin cancer. This research aims to offer a framework to guide future studies and inspire new research directions. In this study, we used the Web of Science Core Collection to collect papers on immunotherapy and melanoma published between 2014 and 2024. With Excel and visualization tools like VOSviewer, COOC 13.2, Citespace, and Bibliometrix (R-Tool of R-Studio), we analyzed the data to spot trends and new focuses in the research. Our findings indicate a substantial surge in research activity concerning immunotherapy and melanoma between 2014 and 2024. The USA and China emerged as leading contributors, engaging in extensive and close collaborative efforts with European counterparts. Furthermore, seven of the top 10 research institutions are located in the USA, with the MD Anderson Cancer Center in Texas being the most productive. In addition, the Journal of Cancer Immunotherapy is the journal with the most articles published in the field. Professor Georgina V. Long from the Melanoma Institute at the University of Sydney was one of the most productive scholars. Keyword analysis shows that immune checkpoint inhibitors, tumor microenvironment and targeted therapies are key areas of interest for the research community. This paper uses bibliometric analysis to outline research trends and key points in immunotherapy and melanoma from 2014 to 2024, which helps understand the current research and guides future research directions.
Collapse
Affiliation(s)
- Zixuan Jia
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Luyan Cao
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, Scotland
| | - Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Almonte AA, Thomas S, Zitvogel L. Microbiota-centered interventions to boost immune checkpoint blockade therapies. J Exp Med 2025; 222:e20250378. [PMID: 40261296 PMCID: PMC12013646 DOI: 10.1084/jem.20250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint blockade therapies have markedly advanced cancer treatment by invigorating antitumor immunity and extending patient survival. However, therapeutic resistance and immune-related toxicities remain major concerns. Emerging evidence indicates that microbial dysbiosis diminishes therapeutic response rates, while a diverse gut ecology and key beneficial taxa correlate with improved treatment outcomes. Therefore, there is a growing understanding that manipulating the gut microbiota could boost therapy efficacy. This review examines burgeoning methods that target the gut microbiome to optimize therapy and innovative diagnostic tools to detect dysbiosis, and highlights challenges that remain to be addressed in the field.
Collapse
Affiliation(s)
- Andrew A. Almonte
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Simon Thomas
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| |
Collapse
|
7
|
Xu DQ, Geng JX, Gao ZK, Fan CY, Zhang BW, Han X, He LQ, Dai L, Gao S, Yang Z, Zhang Y, Arshad M, Fu Y, Mu XQ. To explore the potential combined treatment strategy for colorectal cancer: Inhibition of cancer stem cells and enhancement of intestinal immune microenvironment. Eur J Pharmacol 2025; 998:177533. [PMID: 40120791 DOI: 10.1016/j.ejphar.2025.177533] [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: 10/05/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND The antibiotic salinomycin, a well-known cancer stem cell inhibitor, may impact the diversity of the intestinal microbiota in colorectal cancer (CRC) mice, which plays a pivotal role in shaping the immune system. This study explores the anti-cancer effects and mechanisms of combining salinomycin and fecal microbiota transplantation (FMT) in treating CRC. METHODS FMT was given via enema, while salinomycin was injected intraperitoneally into the CRC mouse model induced by azoxymethane/dextran sodium sulfate. RESULTS In CRC mice, a large number of LGR5-labeled cancer stem cells and severe disturbances in the intestinal microbiota were observed. Interestingly, salinomycin inhibited the proliferation of cancer stem cells without exacerbating the microbial disorder as expected. In comparison to salinomycin treatment, the combination of salinomycin and FMT significantly improved pathological damage and restored intestinal microbial diversity, which is responsible for shaping the anti-cancer immune microenvironment. The supplementation of FMT significantly increased the levels of propionic acid and butyric acid while also promoting the infiltration of CD8+ T cells and Ly6G+ neutrophils, as well as reducing F4/80+ macrophage recruitment. Notably, cytokines that were not impacted by salinomycin exhibited robust reactions to alterations in the gut microbiota. These included pro-inflammatory factors (IL6, IL12b, IL17, and IL22), chemokine-like protein OPN, and immunosuppressive factor PD-L1. CONCLUSIONS Salinomycin plays the role of "eliminating pathogenic qi," targeting cancer stem cells; FMT plays the role of "strengthening vital qi," reversing the intestinal microbiota disorder and enhancing anti-cancer immunity. They have a synergistic effect on the development of CRC.
Collapse
Affiliation(s)
- Dan-Qi Xu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Jia-Xin Geng
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Zhan-Kui Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Chao-Yuan Fan
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Bo-Wen Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Xing Han
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Li-Qian He
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Lin Dai
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Shuo Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Zhou Yang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Yang Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Muhammad Arshad
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Yin Fu
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150006, China.
| | - Xiao-Qin Mu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
8
|
Liu W, Yang X, Zhou Y, Huang Z, Huang J. Gut microbiota in melanoma: Effects and pathogeneses. Microbiol Res 2025; 296:128144. [PMID: 40120565 DOI: 10.1016/j.micres.2025.128144] [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: 11/22/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
The gut microbiota exhibits intricate connections with the body's immune system and holds significant implications for various diseases and cancers. Currently, accumulating evidence suggests a correlation between the composition of the gut microbiota and the development, treatment, and prognosis of melanoma. However, the underlying pathogenesis remains incompletely elucidated. In this comprehensive review, we present an in-depth review of the role played by gut microbiota in melanoma tumorigenesis, growth, metastasis, treatment response, and prognosis. Furthermore, we discuss the potential utility of gut microbiota as a promising prognostic marker. Lastly, we summarize three routes through which gut microbiota influences melanoma: immunity, aging, and the endocrine system. By modulating innate and adaptive immunity in patients with melanoma across different age groups and genders, the gut microbiota plays a crucial role in anti-tumor immune regulation from tumorigenesis to prognosis management, thereby impacting tumor growth and metastasis. This review also addresses current study limitations while highlighting future research prospects.
Collapse
Affiliation(s)
- Wenwen Liu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xin Yang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuwei Zhou
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ziru Huang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jian Huang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan, China.
| |
Collapse
|
9
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
10
|
Sugumar K, Alabd A, Alabd A, Hue JJ, Lyons J, Fields S, Wainberg Z, Zheng L, Coogle B, Kasi A, Grewal N, Kindler HL, Starr J, Sama AR, Winter JM. Exceptional responders to immunotherapy in pancreatic cancer: A multi-institutional case series of a rare occurrence. Oncotarget 2025; 16:427-442. [PMID: 40492845 DOI: 10.18632/oncotarget.28739] [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] [Indexed: 06/12/2025] Open
Abstract
INTRODUCTION Immunotherapy has emerged as a standard treatment option for multiple solid tumors. However, most patients with pancreatic cancer (PC) do not derive a significant benefit. Identification and analyses of exceptional responders could eventually offer hints as to why PC is resistant to immunotherapy. METHODS Oncologists from cancer centers in the United States were contacted to identify patients with PC who responded to immunotherapy. Exceptional responders were defined as those having either partial (PR) or complete response (CR) based on Response Evaluation Criteria in Solid Tumors, or biochemical response (CA 19-9 levels) after starting immunotherapy. Patients receiving concurrent chemotherapy were excluded. RESULTS 14 patients met inclusion criteria. Immunotherapy drugs included checkpoint inhibitors and macrophage inhibitors. Eight patients (42%) were MSI (microsatellite instability)-high. Radiologically, 82% had PR. Four patients (28%) had marked reduction in CA 19-9. The median progression-free survival was 12 months from the start of immunotherapy. Median survival was not reached. The 1- and 2-year survival probabilities were 80%, 70% respectively. CONCLUSION Majority of clinical trials evaluating immunotherapy in PC have yielded disappointing response rates compared to other solid tumors. Our case series adds to published data from early-phase trials supporting the promise of immunotherapy in some patients with PC.
Collapse
Affiliation(s)
- Kavin Sugumar
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Andrew Alabd
- Department of Medicine, Cooper University Healthcare, Camden, NJ 08103, USA
| | - Andre Alabd
- Department of Urology, University of Indiana, Indianapolis, IN 46227, USA
| | - Jonathan J Hue
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Josh Lyons
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Sherri Fields
- Department of Medicine, UCLA/Santa Monica Cancer Center, CA 90404, USA
| | - Zev Wainberg
- Department of Medicine, UCLA/Santa Monica Cancer Center, CA 90404, USA
| | - Lei Zheng
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brianna Coogle
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Anup Kasi
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Nicholas Grewal
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Hedy L Kindler
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jason Starr
- Department of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ashwin R Sama
- Department of Medicine, Jefferson Medical Oncology Associates, Philadelphia, PA 19107, USA
| | - Jordan M Winter
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
11
|
Das M, Kiruthiga C, Shafreen RB, Nachammai K, Selvaraj C, Langeswaran K. Harnessing the human microbiome and its impact on immuno-oncology and nanotechnology for next-generation cancer therapies. Eur J Pharmacol 2025; 996:177436. [PMID: 40023356 DOI: 10.1016/j.ejphar.2025.177436] [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: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The integration of microbiome research and nanotechnology represents a significant advancement in immuno-oncology, potentially improving the effectiveness of cancer immunotherapies. Recent studies highlight the influential role of the human microbiome in modulating immune responses, presenting new opportunities to enhance immune checkpoint inhibitors (ICIs) and other cancer therapies. Nanotechnology offers precise drug delivery and immune modulation capabilities, minimizing off-target effects while maximizing therapeutic outcomes. This review consolidates current knowledge on the interactions between the microbiome and the immune system, emphasizing the microbiome's impact on ICIs, and explores the incorporation of nanotechnology in cancer treatment strategies. Additionally, it provides a forward-looking perspective on the synergistic potential of microbiome modulation and nanotechnology to overcome existing challenges in immuno-oncology. This integrated approach may enhance the personalization and effectiveness of next-generation cancer treatments, paving the way for transformative patient care.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | | | - R Beema Shafreen
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | - Kathiresan Nachammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD Lab, Department of Microbiology, Dr. D. Y. Patil Medical College Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to Be University), Pimpri, Pune, 411018, India.
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India; Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
12
|
Gu BH, Jung HY, Rim CY, Kim TY, Lee SJ, Choi DY, Park HK, Kim M. Comparative Colonisation Ability of Human Faecal Microbiome Transplantation Strategies in Murine Models. Microb Biotechnol 2025; 18:e70173. [PMID: 40448308 DOI: 10.1111/1751-7915.70173] [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: 08/06/2024] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 06/02/2025] Open
Abstract
The gut microbiome plays a crucial role in maintaining intestinal homeostasis and influencing immune-mediated diseases. Human faecal microbiota transplantation (FMT) is often employed in murine models to investigate the role of human microbes in disease regulation, but methods for effective colonisation require refinement. This study aimed to assess the colonisation efficiency of human microbiota in a murine model using FMT with human faeces, focusing particularly on the impact of gut microbiota depletion via polyethylene glycol (PEG) and comparing oral-gastric gavage with enema administration routes. Our findings revealed that PEG-induced depletion enhanced human microbiome colonisation in mice. Oral-gastric gavage prolonged colonisation, while enema administration facilitated quicker resolution of dysbiosis, both inducing selective human microbial colonisation in a time-dependent manner. Notably, genera such as Bacteroides, Blautia, Medicaternibacter and Bifidobacteria were successfully colonised, whereas Roseburia, Anaerostipes, Anaerobutyricum and Faecalibacterium failed to establish in the murine gut post-FMT. These findings highlight the challenges of replicating human gut microbiota in murine models and underscore the importance of selecting appropriate FMT methods based on desired outcomes. This study provides valuable insights into the colonisation dynamics of human microbiota in mice, contributing to the development of more effective FMT strategies for disease treatment.
Collapse
Affiliation(s)
- Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Ho Young Jung
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| | - Chae-Yun Rim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Tae-Yong Kim
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| | - Sang-Jin Lee
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| | - Doo Young Choi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Han-Ki Park
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Myunghoo Kim
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| |
Collapse
|
13
|
Elkrief A, Routy B, Derosa L, Bolte L, Wargo JA, McQuade JL, Zitvogel L. Gut Microbiota in Immuno-Oncology: A Practical Guide for Medical Oncologists With a Focus on Antibiotics Stewardship. Am Soc Clin Oncol Educ Book 2025; 45:e472902. [PMID: 40262063 DOI: 10.1200/edbk-25-472902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The gut microbiota has emerged as a critical determinant of immune checkpoint inhibitor (ICI) efficacy, resistance, and toxicity. Retrospective and prospective studies profiling the taxonomic composition of intestinal microbes of patients treated with ICI have revealed specific gut microbial signatures associated with response. By contrast, dysbiosis, which can be caused by chronic inflammatory processes (such as cancer) or comedications, is a risk factor of resistance to ICI. Recent large-scale meta-analyses have confirmed that antibiotic (ATB) use before or during ICI therapy alters the microbiota repertoire and significantly shortens overall survival, even after adjusting for prognostic factors. These results underscore the importance of implementing ATB stewardship recommendations in routine oncology practice. Microbiota-centered interventions are now being explored to treat gut dysbiosis and optimize ICI responses. Early-phase clinical trials evaluating fecal microbiota transplantation (FMT) from ICI responders or healthy donors have shown that this approach is safe and provided preliminary data on potential efficacy to overcome both primary and secondary resistance to ICI in melanoma, non-small cell lung cancer, and renal cell carcinoma. More targeted interventions including live bacterial products including Clostridium butyricum and Akkermansia massiliensis represent novel microbiome-based adjunct therapies. Likewise, dietary interventions, such as high-fiber diets, have shown promise in enhancing ICI activity. In this ASCO Educational Book, we summarize the current state-of-the-evidence of the clinical relevance of the intestinal microbiota in cancer immunotherapy and provide a practical guide for ATB stewardship.
Collapse
Affiliation(s)
- Arielle Elkrief
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Bertrand Routy
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
| | - Laura Bolte
- Department of Medical Oncology, University Groningen and University Medical Center, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University Groningen and University Medical Center, Groningen, the Netherlands
| | | | | | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|
14
|
Silveira MAD, Rodrigues RR, Trinchieri G. Intestinal Microbiome Modulation of Therapeutic Efficacy of Cancer Immunotherapy. Gastroenterol Clin North Am 2025; 54:295-315. [PMID: 40348489 PMCID: PMC12066836 DOI: 10.1016/j.gtc.2024.10.005] [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] [Indexed: 05/14/2025]
Abstract
Bacteria are associated with certain cancers and may induce genetic instability and cancer progression. The gut microbiome modulates the response to cancer therapy. Training machine learning models with response associated taxa or bacterial genes predict patients' response to immunotherapies with moderate accuracy. Clinical trials targeting the gut microbiome to improve immunotherapy efficacy have been conducted. While single bacterial strains or small consortia have not be reported yet to be successful, encouraging results have been reported in small single arm and randomized studies using transplant of fecal microbiome from cancer patients who successfully responded to therapy or from healthy volunteers.
Collapse
Affiliation(s)
- Maruhen A D Silveira
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4146, Bethesda, MD 20852, USA
| | - Richard R Rodrigues
- Microbiome and Genetics Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4140B, Bethesda, MD 20852, USA; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Giorgio Trinchieri
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4146, Bethesda, MD 20852, USA.
| |
Collapse
|
15
|
Wang C, Dong T, Rong X, Yang Y, Mou J, Li J, Ge J, Mu X, Jiang J. Microbiome in prostate cancer: pathogenic mechanisms, multi-omics diagnostics, and synergistic therapies. J Cancer Res Clin Oncol 2025; 151:178. [PMID: 40450182 DOI: 10.1007/s00432-025-06187-w] [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: 10/01/2024] [Accepted: 03/31/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Prostate cancer (PCa) is a leading cause of cancer-related deaths in men, with the microbiome emerging as a significant factor in its development and progression. Understanding the microbiome's role could provide new insights into PCa pathogenesis and treatment. OBJECTIVE This review aims to explore the interactions between the microbiome and PCa, focusing on microbial imbalances and their effects on immune responses, inflammation, and hormone levels. It also discusses advanced research techniques and the potential for microbiome modulation in PCa management. METHODS The review synthesizes current literature on the microbiome's role in PCa, highlighting differences in microbial composition between cancerous and healthy prostate tissues. It examines techniques such as high-throughput sequencing and metagenomics and explores the mechanisms through which the microbiome influences PCa. CONCLUSIONS The review reveals substantial microbial differences in prostate tissues of PCa patients compared to healthy individuals, indicating a potential link between microbiome alterations and disease progression. It highlights the promise of microbiome-based strategies for diagnosis and treatment and underscores the need for further research into personalized, microbiome-centric approaches for PCa management.
Collapse
Affiliation(s)
- Chengran Wang
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Tianqi Dong
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Xin'ao Rong
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Yuce Yang
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Jianhui Mou
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Jiaqi Li
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Jianli Ge
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Xupeng Mu
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China.
| | - Jinlan Jiang
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China.
| |
Collapse
|
16
|
Torabi S, Nahidi Y, Ghasemi SZ, Reihani A, Samadi A, Ramezanghorbani N, Nazari E, Davoudi S. Evaluation of skin cancer prevention properties of probiotics. GENES & NUTRITION 2025; 20:12. [PMID: 40410666 PMCID: PMC12101031 DOI: 10.1186/s12263-025-00770-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 05/04/2025] [Indexed: 05/25/2025]
Abstract
Bacteria play a crucial role in human health and disease pathogenesis. In recent years, the therapeutic potential of probiotics has gained increasing attention, with studies suggesting their application in treating various diseases, including cancer. We evaluated clinical data supporting the use of oral and topical probiotics for skin malignancies by conducting a literature search in PubMed and Google Scholar. Although limited, clinical trials investigating probiotics in cancer prevention and treatment have shown promising results, particularly in controlling tumor progression and enhancing therapeutic outcomes. Emerging research suggests that probiotics may contribute to skin cancer prevention by modulating the gut and skin microbiomes, enhancing immune responses, exerting antioxidant and anti-inflammatory effects, and inducing apoptosis. Given their antiproliferative and pro-apoptotic effects on carcinoma cells, probiotic-based therapies may serve as potential cancer-preventive agents and adjunctive treatments during conventional therapies. Key findings from our review highlight the ability of probiotics to influence cancer progression through immune regulation, apoptosis induction, and modulation of inflammatory pathways. However, further well-designed clinical trials are needed to validate these findings and establish probiotics as a viable therapeutic approach in oncology.
Collapse
Affiliation(s)
- Shatila Torabi
- Department of Dermatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Cutaneous Leishmaniasis Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yalda Nahidi
- Department of Dermatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Cutaneous Leishmaniasis Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Zahra Ghasemi
- Cutaneous Leishmaniasis Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirali Reihani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Samadi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Nahid Ramezanghorbani
- Assistant Professor, Department of Development & Coordination Scientific Information and Publications, Deputy of Research & Technology, Ministry of Health & Medical Education, Tehran, Iran
| | - Elham Nazari
- Proteomics Research Center, System Biology Institute, Faculty of Paramedical sciences, Shahid Beheshti university of medical sciences, Tehran, Iran
| | - Sima Davoudi
- Department of Dermatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Cutaneous Leishmaniasis Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Wekking D, Silva CAC, Viscò R, Denaro N, Lambertini M, Maccioni A, Loddo E, Willard-Gallo K, Scartozzi M, Derosa L, Solinas C. The interplay between gut microbiota, antibiotics, and immune checkpoint inhibitors in patients with cancer: A narrative review with biological and clinical aspects. Crit Rev Oncol Hematol 2025; 212:104767. [PMID: 40414545 DOI: 10.1016/j.critrevonc.2025.104767] [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: 04/05/2025] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting the programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen 4 (CTLA-4) pathways have revolutionized cancer therapy. However, primary and secondary resistance to ICI pose significant challenges. Recent studies underscore the critical role of gut microbiota (GM) in modulating ICI efficacy by shaping host immune responses and regulating the tumor microenvironment (TME). The composition of the GM has been linked to ICI treatment outcomes, with certain microbial taxa, such as Faecalibacterium spp., Bifidobacterium spp., Eubacterium spp., Roseburia spp., and Akkermansia muciniphila, associated with favorable responses. Mechanistically, the GM affects immune responses via multiple pathways, including induction of T cell differentiation, promotion of anti- or proinflammatory cytokine environments, and enhancement of T cell priming and effector functions. Moreover, microbial-derived metabolites play a role in shaping tumor immune responses and influencing ICI efficacy. Antibiotic treatment can disrupt GM diversity and composition (gut dysbiosis), potentially diminishing ICI effectiveness. A deeper understanding of the interplay between GM, antibiotic treatment, and ICI efficacy is crucial for developing personalized therapeutic strategies to improve patient outcomes. Herein, we review current evidence on the association between specific microbial taxa and tumor immunosurveillance, the impact of antibiotics on the GM composition and immune modulation, and its implications for ICI therapy efficacy.
Collapse
Affiliation(s)
- Demi Wekking
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Roberto Viscò
- Ospedale Sant'Antonio Abate, Patologica Clinica, ASP Trapani, Italy
| | - Nerina Denaro
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Antonio Maccioni
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato (CA) Italy
| | - Erica Loddo
- Gastroenterology University Hospital, Cagliari, Italy
| | | | - Mario Scartozzi
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato (CA) Italy; University Hospital of Cagliari, Italy
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Cinzia Solinas
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato (CA) Italy
| |
Collapse
|
18
|
Hu X, Jiang T, Wang J. Molecular subtype characteristics and development of prognostic model based on inflammation-related gene in lung adenocarcinoma. Discov Oncol 2025; 16:875. [PMID: 40407957 PMCID: PMC12102027 DOI: 10.1007/s12672-025-02513-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Abstract
As one of the leading causes of death worldwide, lung adenocarcinoma (LUAD) currently lacks satisfactory treatment outcomes. The inflammatory process, closely associated with the formation of the tumor microenvironment and immune evasion, plays a crucial role in LUAD development. This study utilized data from public databases to analyze inflammation-related genes (INF) associated with prognosis in LUAD. Based on differentially expressed INF, molecular subtypes of LUAD were identified. Subsequently, a novel INF scoring system was developed to establish a prognostic model for LUAD patients, assessing its independence and reliability. Comprehensive evaluations, including immune microenvironment infiltration features, somatic mutation characteristics, and differences in immune therapy responsiveness, were conducted to characterize the prognostic model associated with INF. We further selected MMP14 from the screened INF targets for further in vitro experiments. Experiments such as western blot, qRT-PCR, colony-forming assay and Transwell assay confirmed that downregulation of MMP14 could inhibit the cloning, proliferation and invasion of lung cancer cells, thus confirming the results of bioinformatics. Our findings provide evidence from a new perspective on the role of inflammation in LUAD and offer new insights for clinical precision and personalized therapy.
Collapse
Affiliation(s)
- Xuelei Hu
- Department of Thoracic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Tengfei Jiang
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Jinxiang Wang
- Department of Respiratory and Critical Care Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China.
| |
Collapse
|
19
|
Heyer R, Wolf M, Benndorf D, Uzzau S, Seifert J, Grenga L, Pabst M, Schmitt H, Mesuere B, Van Den Bossche T, Haange SB, Jehmlich N, Di Luca M, Ferrer M, Serrano-Villar S, Armengaud J, Bode HB, Hellwig P, Masselot CR, Léonard R, Wilmes P. Metaproteomics in the One Health framework for unraveling microbial effectors in microbiomes. MICROBIOME 2025; 13:134. [PMID: 40410872 PMCID: PMC12100821 DOI: 10.1186/s40168-025-02119-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/21/2025] [Indexed: 05/25/2025]
Abstract
One Health seeks to integrate and balance the health of humans, animals, and environmental systems, which are intricately linked through microbiomes. These microbial communities exchange microbes and genes, influencing not only human and animal health but also key environmental, agricultural, and biotechnological processes. Preventing the emergence of pathogens as well as monitoring and controlling the composition of microbiomes through microbial effectors including virulence factors, toxins, antibiotics, non-ribosomal peptides, and viruses holds transformative potential. However, the mechanisms by which these microbial effectors shape microbiomes and their broader functional consequences for host and ecosystem health remain poorly understood. Metaproteomics offers a novel methodological framework as it provides insights into microbial dynamics by quantifying microbial biomass composition, metabolic functions, and detecting effectors like viruses, antimicrobial resistance proteins, and non-ribosomal peptides. Here, we highlight the potential of metaproteomics in elucidating microbial effectors and their impact on microbiomes and discuss their potential for modulating microbiomes to foster desired functions.
Collapse
Affiliation(s)
- Robert Heyer
- Multidimensional Omics Analyses Group, Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Straße 11, 44139, Dortmund, Germany.
- Multidimensional Omics Analyses Group, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany.
| | - Maximilian Wolf
- Multidimensional Omics Analyses Group, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Dirk Benndorf
- Bioprocess Engineering, Otto-Von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems Magdeburg, Sandtorstraße 1, 39106, Magdeburg, Germany
- Applied Biosciences and Process Engineering, Anhalt University of Applied Sciences, Köthen, Germany
| | - Sergio Uzzau
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Jana Seifert
- Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str, Stuttgart, Germany
- HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Leonore-Blosser-Reisen Weg, Stuttgart, Germany
| | - Lucia Grenga
- Département Médicaments Et Technologies Pour La Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-Sur-Cèze, France
| | - Martin Pabst
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands
| | - Heike Schmitt
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands
- Institute for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Bart Mesuere
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000, Ghent, Belgium
| | - Tim Van Den Bossche
- VIB - UGent Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9052, Ghent, Belgium
| | - Sven-Bastiaan Haange
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ GmbH, Permoserstrasse 15, 04318, Leipzig, Germany
| | - Nico Jehmlich
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ GmbH, Permoserstrasse 15, 04318, Leipzig, Germany
| | | | - Manuel Ferrer
- Instituto de Catalisis y Petroleoquimica (ICP), CSIC, 28049, Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Hospital Universitario Ramon y Cajal, Instituto de Investigación Sanitaria Ramón y Cajal (IRYCIS), CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Jean Armengaud
- Département Médicaments Et Technologies Pour La Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-Sur-Cèze, France
| | - Helge B Bode
- Department of Natural Products in Organismic Interactions, Max-Planck-Institut for Terrestrial Microbiology, Karl-Von-Frisch-Str. 10, 35043, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Phillips University Marburg, 35043, Marburg, Germany
- Department of Chemistry, Phillips University Marburg, 35043, Marburg, Germany
| | - Patrick Hellwig
- Bioprocess Engineering, Otto-Von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106, Magdeburg, Germany
| | | | - Renaud Léonard
- Université de Lille, CNRS, UMR, 8576 - UGSF, Lille, France
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-Sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, L-4362, Esch-Sur-Alzette, Luxembourg
| |
Collapse
|
20
|
Montanari E, Bernardo G, Le Noci V, Anselmi M, Pupa SM, Tagliabue E, Sommariva M, Sfondrini L. Biofilm formation by the host microbiota: a protective shield against immunity and its implication in cancer. Mol Cancer 2025; 24:148. [PMID: 40399923 PMCID: PMC12093748 DOI: 10.1186/s12943-025-02348-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025] Open
Abstract
Human-resident microbes typically cluster into biofilms - structurally organized communities embedded within a matrix of self-produced extracellular polymeric substance (EPS) that serves as a protective shield. These biofilms enhance microbial survival and functional adaptability, favoring a symbiotic relationship with the host under physiological conditions. However, biofilms exhibit a dual role in modulating the immune response. If their ability to promote tolerance is key to safeguarding homeostasis, by contrast, their persistence can overcome the cutting-edge balance resulting in immune evasion, chronic inflammation and development of numerous diseases such as cancer. Recent evidence highlights the significance of cancer-associated microbiota in shaping the tumor microenvironment (TME). These microbial inhabitants often exhibit biofilm-like structures, which may protect them from host immune responses and therapeutic interventions. The presence of biofilm-forming microbiota within the TME may promote chronic inflammation, and release of bioactive molecules that interfere with immune surveillance mechanisms, thereby enabling cancer cells to evade immune destruction. This review delves into the complex interplay between biofilms and cancer, with particular focus on the tumor-associated microbiota and the implications of biofilm involvement in modulating the immune landscape of the TME. Addressing this intricate relationship holds promises for innovative therapeutic approaches aimed at reprogramming the microbiota-cancer axis for better clinical outcomes.
Collapse
Affiliation(s)
- Elena Montanari
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Valentino Le Noci
- Department of Experimental Oncology, Microenvironment and Biomarkers of Solid Tumors Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Martina Anselmi
- Department of Experimental Oncology, Microenvironment and Biomarkers of Solid Tumors Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Serenella M Pupa
- Department of Experimental Oncology, Microenvironment and Biomarkers of Solid Tumors Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elda Tagliabue
- Department of Experimental Oncology, Microenvironment and Biomarkers of Solid Tumors Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- Department of Experimental Oncology, Microenvironment and Biomarkers of Solid Tumors Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy.
- Department of Experimental Oncology, Microenvironment and Biomarkers of Solid Tumors Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| |
Collapse
|
21
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [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: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Peng Y, Tang Q, Wu S, Zhao C. Associations of Atopobium, Garderella, Megasphaera, Prevotella, Sneathia, and Streptococcus with human papillomavirus infection, cervical intraepithelial neoplasia, and cancer: a systematic review and meta-analysis. BMC Infect Dis 2025; 25:708. [PMID: 40380083 PMCID: PMC12082921 DOI: 10.1186/s12879-025-10851-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/24/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Lactobacillus spp. depleted and high diversity of vaginal microbiota is closely related to human papillomavirus infection and cervical cancer. However, the role of other microbial communities in human papillomavirus infection and cervical cancer is still unclear. OBJECTIVE This study aims to systematically review the existing literature and perform a meta-analysis to statistically evaluate the relationship between vaginal microbiota, human papillomavirus infection, cervical intraepithelial neoplasia, and cervical cancer at the genus level. METHODS A comprehensive search of PubMed, Web of Science, and Embase databases was conducted to identify relevant studies. We synthesized data on the relative abundance of specific bacterial species associated with human papillomavirus status and cervical lesions. SPSS 25.0 was used to compare relative abundance among multiple groups. RESULTS The meta-analysis included 17 observational studies published between 2019 and 2023, involving 2014 participants from Asia, North America, and Africa. We found that specific vaginal microorganisms, such as Gardnerella, Prevotella, Sneathia, and Streptococcus, showed increased relative abundance with the severity of cervical lesions in human papillomavirus-negative, human papillomavirus-positive, cervical intraepithelial neoplasia, and cervical cancer patients. However, no statistically significant differences were found in that regard. Notably, Prevotella was significantly more abundant in cervical cancer patients compared to human papillomavirus-negative individuals. Sneathia was also found to be more abundant in cervical intraepithelial neoplasia and cervical cancer patients. CONCLUSIONS The specific vaginal microbial species are associated with human papillomavirus infection status and the severity of cervical lesions that may have significant implications for the prevention and treatment strategies of cervical cancer.
Collapse
Affiliation(s)
- Yan Peng
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Longshan Road 120, Yubei District, Chongqing, 400010, People's Republic of China
| | - Qin Tang
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Longshan Road 120, Yubei District, Chongqing, 400010, People's Republic of China
| | - Shiming Wu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Longshan Road 120, Yubei District, Chongqing, 400010, People's Republic of China
| | - Chengzhi Zhao
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Longshan Road 120, Yubei District, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
23
|
Hesami Z, Sabzehali F, Khorsand B, Alipour S, Sadeghi A, Asri N, Pazienza V, Houri H. Microbiota as a state-of-the-art approach in precision medicine for pancreatic cancer management: A comprehensive systematic review. iScience 2025; 28:112314. [PMID: 40276756 PMCID: PMC12019022 DOI: 10.1016/j.isci.2025.112314] [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: 10/27/2024] [Revised: 12/22/2024] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Emerging evidence suggests that harnessing the microbiome holds promise for innovative diagnostic and therapeutic strategies in the management of pancreatic cancer (PC). This study aims to systematically summarize the microbial markers associated with PC and assess their potential application in clinical outcome. Forty-one studies were included to assess the associations between microbial markers and PC. Among these, 13 were developed prediction models related to the microbiome in which the highest diagnostic and prognostic model belong to blood and intratumor markers, respectively. Notably, findings that utilize microbiotas from various body sites were elucidated, demonstrating their importance as unique signatures in biomarker discovery for diverse clinical applications. This review provides unique perspectives on overcoming challenges in PC by highlighting potential microbial-related markers as non-invasive approaches. Further clinical studies should evaluate the utility and accuracy of key indicators in the microbiome as a personalized tool for managing PC.
Collapse
Affiliation(s)
- Zeinab Hesami
- Student Research Committee, Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fattaneh Sabzehali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Samira Alipour
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nastaran Asri
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Hamidreza Houri
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Amen RA, Hassan YM, Essmat RA, Ahmed RH, Azab MM, Shehata NR, Elgazzar MM, El-Sayed WM. Harnessing the Microbiome: CRISPR-Based Gene Editing and Antimicrobial Peptides in Combating Antibiotic Resistance and Cancer. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10573-8. [PMID: 40377870 DOI: 10.1007/s12602-025-10573-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
The growing crisis of antibiotic resistance and the increasing incidence of cancer have prompted the exploration of innovative approaches, such as gene editing and antimicrobial peptides (AMPs). The human microbiome is integral to various aspects of health, disease, and therapeutic development, influencing metabolic pathways, immune function, and pathogen resistance. Recent advances in gene editing technologies, particularly CRISPR (clustered regularly interspaced short palindromic repeats), have opened new avenues for leveraging the microbiome to address complex medical challenges, including combating multidrug-resistant pathogens and cancer. The microbiome plays a crucial role in combating antibiotic resistance by modulating microbial communities, influencing pathogen survival and susceptibility to treatments. This review explores the microbiome's dynamic role in metabolic regulation, its contribution to cancer management, and how AMPs help maintain homeostasis and exhibit emerging anticancer properties, supported by both preclinical findings and clinical evidence. Additionally, CRISPR-based microbiome engineering offers potential to enhance host-microbiome interactions, optimizing therapeutic outcomes. The integration of microbiome metagenomics and proteomics has led to the discovery of novel AMPs with targeted anticancer effects. Innovative strategies, such as engineered probiotics and CRISPR-based microbiome engineering, present exciting prospects for next-generation therapies. Despite these advances, the translation of microbiome-based therapies into clinical settings remains challenging due to ethical, regulatory, and ecological hurdles. This review underscores the transformative potential of microbiome-based interventions, emphasizing the role of personalized medicine in maximizing therapeutic efficacy. Furthermore, we also address critical research gaps, limitations, and future directions, including optimizing AMP stability, delivery, and bioavailability, as well as overcoming the regulatory and ethical challenges in clinical translation.
Collapse
Affiliation(s)
- Radwa A Amen
- Department of Biotechnology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Yaser M Hassan
- Biotechnology Program, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Rawan A Essmat
- Faculty of Pharmacy, Modern University for Information and Technology, Cairo, 11728, Egypt
| | - Rana H Ahmed
- Biotechnology Program, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Marwan M Azab
- Molecular Biotechnology Program, Faculty of Science, Helwan University, Ain Helwan, Cairo, Egypt
| | - Nadia R Shehata
- College of Biotechnology, Misr University for Science and Technology, Giza, 12596, Egypt
| | | | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia 11566, Cairo, Egypt.
| |
Collapse
|
25
|
Zhang X, Fam KT, Dai T, Hang HC. Microbiota mechanisms in cancer progression and therapy. Cell Chem Biol 2025; 32:653-677. [PMID: 40334660 DOI: 10.1016/j.chembiol.2025.04.005] [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: 11/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
The composition of the microbiota in patients has been shown to correlate with cancer progression and response to therapy, highlighting unique opportunities to improve patient outcomes. In this review, we discuss the challenges and advancements in understanding the chemical mechanisms of specific microbiota species, pathways, and molecules involved in cancer progression and treatment. We also describe the modulation of cancer and immunotherapy by the microbiota, along with approaches for investigating microbiota enzymes and metabolites. Elucidating these specific microbiota mechanisms and molecules should offer new opportunities for developing enhanced diagnostics and therapeutics to improve outcomes for cancer patients. Nonetheless, many microbiota mechanisms remain to be determined and require innovative chemical genetic approaches.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tingting Dai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
26
|
Liu S, Pan Y, Zheng C, Zheng Q, Du Y, Zheng Y, Tang H, Liu X, Mou J, Zeng X, Yang Z, Gui W, Tang Y, Xu M, Ye Z, Su H, Xu Q, Meng X. Tumor-colonizing Pseudoalteromonas elyakovii metabolically reprograms the tumor microenvironment and promotes breast ductal carcinoma. mBio 2025; 16:e0387324. [PMID: 40192290 PMCID: PMC12077203 DOI: 10.1128/mbio.03873-24] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/03/2025] [Indexed: 05/15/2025] Open
Abstract
The correlation between the microbiota found in tumors and tumor development is being progressively understood, specifically regarding its involvement in the initiation and advancement of tumors. We examined a total of 102 samples, examining the microbial composition at the species level in each person unveiled significant variations in both the microbial makeup and tumor proportions among individuals, examining the fluctuating alterations in the microbial profile during breast cancer advancement and progression. The levels of expression for Pseudoalteromonas elyakovii were notably elevated in the tumor groups when compared to the para-cancer normal group, aligning with the results obtained from qRT-PCR analysis. The relationship between tumor immunity and microorganisms within the tumor was investigated using double immunofluorescence staining combined with SweAMI probe in situ hybridization and scRNA-seq, allowing for an in-depth analysis of intratumoral microorganisms. Experiments have demonstrated that the supernatant derived from P. elyakovii displayed a significant ability to promote tumor growth and stimulation. In summary, we describe the characteristics of the intratumoral microbiota and the tumor-promoting effects of P. elyakovii supernatant within a small dose range in ductal carcinoma of the breast and characterize the potential clinical application value of intratumoural microorganisms in the progression of cancer and immunotherapy. IMPORTANCE Despite the existing studies, the specific microbial factors that influence the occurrence and progression of breast cancer still remain unclear. Researchers have clarified the distinctive microbial profile related to ductal carcinoma, a common histological type of breast cancer, in order to identify tumor-specific microbes and their roles in tumorigenesis. With the tumor microbiome as the focus, the enrichment of Pseudoalteromonas elyakovii features accelerates the disease progression in patients with ductal carcinoma of the breast. This study reveals the initial role relationship and innovative findings between Pseudoalteromonas elyakovii and ductal carcinoma in the breast.
Collapse
Affiliation(s)
- Shuyan Liu
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Youpeng Pan
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chaopeng Zheng
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qinghui Zheng
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
| | - Yaoqiang Du
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yajuan Zheng
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
| | - Hongchao Tang
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
| | - Xiaozhen Liu
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
| | - Jiancheng Mou
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xin Zeng
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
| | - Zhuotao Yang
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wenjuan Gui
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yuning Tang
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mingxing Xu
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhihao Ye
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Haotian Su
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
- The Second School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qiuran Xu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xuli Meng
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema and Stasis of Breast Cancer, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Barroso-Sousa R, Zanudo JGT, Li T, Reddy SM, Emens LA, Kuntz TM, Silva CAC, AlDubayan SH, Chu H, Overmoyer B, Lange P, DiLullo MK, Montesion M, Kasparian J, Hughes ME, Attaya V, Basta A, Lin NU, Tayob N, Jeselsohn R, Mittendorf EA, Tolaney SM. Nivolumab plus low-dose ipilimumab in hypermutated HER2-negative metastatic breast cancer: a phase II trial (NIMBUS). Nat Commun 2025; 16:4430. [PMID: 40360544 PMCID: PMC12075640 DOI: 10.1038/s41467-025-59695-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
In the phase II NIMBUS trial, patients with human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC) and high tumor mutational burden (TMB ≥ 9 mut/Mb) received nivolumab (3 mg/kg biweekly) and low-dose ipilimumab (1 mg/kg every 6 weeks) for 2 years or until progression. The primary endpoint was objective response rate (ORR) per RECIST 1.1 criteria. Among 30 patients enrolled, the median TMB was 10.9 mut/Mb (range: 9-110) and the confirmed objective response rate was 20%. Secondary endpoints included progression-free survival, overall survival, clinical benefit rate, and safety and tolerability, including immune-related adverse events (irAEs). A prespecified correlative outcome was to evaluate the ORR in patients with a TMB ≥ 14 mut/Mb. Patients with TMB ≥ 14 mut/Mb (n = 6) experienced higher response rates (60% vs 12%; p = 0.041) and showed a trend towards improved progression-free survival and overall survival compared to patients with TMB < 14 mut/Mb. Exploratory genomic analyses suggested that ESR1 and PTEN mutations may be associated with poor response, while clinical benefit was associated with a decrease or no change in tumor fraction by serial circulating tumor DNA during treatment. Stool microbiome analysis revealed that baseline blood TMB, PD-L1 positivity, and immune-related diarrhea are associated with distinct taxonomic profiles. In summary, some patients with hypermutated HER2-negative MBC experience extended clinical benefit with a dual immunotherapy regimen; a higher TMB, and additional genomic and microbiome biomarkers may optimize patient selection for therapy with nivolumab plus low-dose ipilimumab. (Funded by Bristol Myers Squibb; ClinicalTrials.gov identifier, NCT03789110).
Collapse
Affiliation(s)
| | - Jorge Gomez Tejeda Zanudo
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tianyu Li
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Leisha A Emens
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Thomas M Kuntz
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Hoyin Chu
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Beth Overmoyer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paulina Lange
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Molly K DiLullo
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | | | - Julie Kasparian
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Melissa E Hughes
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Victoria Attaya
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Ameer Basta
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nabihah Tayob
- Harvard Medical School, Boston, MA, USA
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rinath Jeselsohn
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Fernandez E, Wargo JA, Helmink BA. The Microbiome and Cancer: A Translational Science Review. JAMA 2025:2833859. [PMID: 40354071 DOI: 10.1001/jama.2025.2191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Importance Growing evidence suggests that microbes located within the gastrointestinal tract and other anatomical locations influence the development and progression of diseases such as cancer. Observations Clinical and preclinical evidence suggests that microbes in the gastrointestinal tract and other anatomical locations, such as the respiratory tract, may affect carcinogenesis, development of metastases, cancer treatment response, and cancer treatment-related adverse effects. Within tumors of patients with cancer, microbes may affect response to treatment, and therapies that reduce or eliminate these microbes may improve outcomes in patients with cancer. Modulating gastrointestinal tract (gut) microbes through fecal microbiota transplant and other strategies such as dietary intervention (eg, high-fiber diet intervention) has improved outcomes in small studies of patients treated with cancer immunotherapy. In contrast, disruption of the gut microbiota by receipt of broad-spectrum antibiotics prior to treatment with cancer immunotherapy has been associated with poorer overall survival and higher rates of adverse effects in patients treated with immune checkpoint blockade for solid tumors and also with chimeric antigen receptor T-cell therapy for hematologic malignancies. Conclusions and Relevance Microbes in the gut and other locations in the body may influence the development and progression of cancer and may affect the response to adverse effects from cancer therapy. Future therapies targeting microbes in the gut and other locations in the body could potentially improve outcomes in patients with cancer.
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Beth A Helmink
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
29
|
Wang Y, Wang X, Li W, Huang H, Di C, Yang Q. Huaier enhances the antitumor effects of CDK 4/6 inhibitor by remodeling the immune microenvironment and gut microbiota in breast cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119723. [PMID: 40179997 DOI: 10.1016/j.jep.2025.119723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/01/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Trametes robiniophila Murr (Huaier), a traditional Chinese medicine (TCM), has shown promising effects as a complementary therapy in the treatment of multiple kinds of cancers. AIM OF THE STUDY This study aimed to systematically evaluate whether Huaier could enhance the antitumor effects of palbociclib, a highly selective CDK4/6 inhibitor. MATERIALS AND METHODS A breast cancer xenograft mouse model was constructed to assess the anticancer effects of Huaier and palbociclib. Transcriptomic analysis was performed to identify the differentially expressed genes and associated pathways. Flow cytometry, immunofluorescence and immunohistochemical staining were then conducted to observe the infiltration of immune cells in the tumor microenvironment (TME). The composition of the gut microbiota was explored through 16S rDNA sequencing, and a pseudosterile mouse model was established to verify the contribution of the gut microbiota to the antitumor effects of Huaier and palbociclib. RESULTS Oral administration of Huaier enhanced the antitumor effects of palbociclib in breast cancer. Transcriptomic analysis of tumor tissues revealed that the differentially expressed genes were related to immune functions. Indeed, the addition of Huaier to palbociclib therapy further increased the ratio of CD8+/CD3+ T cells and the abundance of GzmB+CD8+ T cells in the TME. Mechanistically, sequencing of 16S rDNA revealed that Huaier reshaped the gut microbiota composition in favor of Akkermansia, which positively modulated antitumor immune responses. The pseudosterile mouse model verified the necessity of the gut microbiota for the synergistic effect of Huaier on palbociclib. Furthermore, butyrate, a metabolite of Akkermansia, synergized with palbociclib to suppress tumor progression and promote the infiltration of CD8+ T cells in the TME. CONCLUSIONS Our data suggested that Huaier augmented the antitumor effects of palbociclib by increasing the abundance of Akkermansia in gut microbiota, which contributed to the enhancement of anti-tumor immunity. Consequently, combining palbociclib with Huaier may serve as a promising strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Xiaolong Wang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Wenhao Li
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Hanwen Huang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Chenyu Di
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China; Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, China; Research Institute of Breast Cancer, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
30
|
Jin Y, Jie Z, Fan X. Gut microbes and immunotherapy for non-small cell lung cancer: a systematic review. Front Oncol 2025; 15:1518474. [PMID: 40406244 PMCID: PMC12095033 DOI: 10.3389/fonc.2025.1518474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Emerging evidence underscores gut microbiota's role in modulating lung cancer immunotherapy outcomes, though specific impacts on immune checkpoint inhibitors (ICIs) and associated adverse events (AEs) require further clarity. This review synthesizes findings from 15 studies examining gut microbiota-ICI interactions in non-small cell lung cancer (NSCLC), alongside studies investigating antibiotics, proton pump inhibitors (PPIs), probiotics, and diet as modulating factors. Results indicate that Actinobacteria, Bacteroides, and Verrucomicrobiota correlate with positive ICI responses, while Bacillota shows variable associations; notably, Bacillota-enriched patients had fewer immunotherapy-related AEs. The administration of antibiotics and PPIs within a month before ICIs was linked to diminished efficacy, whereas probiotics correlated with enhanced outcomes. Plant-based diets are also aligned with dietary patterns supportive of ICIs. These findings suggest that analyzing gut microbiota composition could improve the ability to predict NSCLC patient responses to ICIs. Additionally, judicious use of antibiotics, PPIs, probiotics, and dietary adjustments may optimize immunotherapy outcomes and mitigate adverse effects.
Collapse
Affiliation(s)
- Yali Jin
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhiqian Jie
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
31
|
Glapiński F, Zając W, Fudalej M, Deptała A, Czerw A, Sygit K, Kozłowski R, Badowska-Kozakiewicz A. The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies. Cancers (Basel) 2025; 17:1599. [PMID: 40427098 PMCID: PMC12110676 DOI: 10.3390/cancers17101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer (PC), with pancreatic ductal adenocarcinoma (PDAC) comprising about 90% of all cases, is one of the most aggressive and lethal solid tumors. PDAC remains one of the most significant challenges of oncology to this day due to its inadequate response to conventional treatment, gradual rise in incidence since 2004, and poor five-year survival rates. As cancer cells are the primary adversary in this uneven fight, they remain the primary research target. Nevertheless, increasing attention is being paid to the tumor microenvironment (TME). The most crucial TME constellation components are immune cells, especially macrophages, stellate cells and lymphocytes, fibroblasts, bacterial and fungal microflora, and neuronal cells. Depending on the particular phenotype of these cells, the composition of the microenvironment, and the cell ratio, patients can experience different disease outcomes and varying vulnerability to treatment approaches. This study aims to present the current knowledge and review the most up-to-date scientific findings regarding the microenvironment of PC. It contains detailed information on the structure and cellular composition of the stroma, including its impact on disease development, metastasis, and response to treatment, as well as the therapeutic opportunities that arise from targeting this tissue.
Collapse
Affiliation(s)
- Franciszek Glapiński
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Weronika Zając
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Marta Fudalej
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
- Department of Oncology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Andrzej Deptała
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Department of Economic and System Analyses, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Remigiusz Kozłowski
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| | - Anna Badowska-Kozakiewicz
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
| |
Collapse
|
32
|
von Bubnoff D, Schmitt C, Goldinger SM, Schadendorf D, Kähler KC, Hafner C, Kramer N, Fröhlich W, Dummer R, Berking C, Schliep S, Kirchberger MC, Heinzerling L. Prognostic and predictive value of IDO expression in metastatic melanoma treated with Ipilimumab. PLoS One 2025; 20:e0321937. [PMID: 40334245 PMCID: PMC12058187 DOI: 10.1371/journal.pone.0321937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 03/11/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The tumor microenvironment is crucial for prognosis and response to immunotherapy in several tumor entities. METHODS In a multicenter retrospective study, a total of 86 tumor samples from patients with metastatic melanoma were evaluated for baseline expression of indoleamine 2,3-dioxygenase (IDO) and programmed death ligand 1 (PD-L1). Expression patterns of IDO and PD-L1 on tumor cells and antigen-presenting cells (APCs) as determined by immunohistochemical (IHC) staining of paraffin-embedded tissue sections were correlated with response to ipilimumab and overall survival (OS). Statistical analysis was performed using the Spearman correlation, the Mann-Whitney test and Kaplan-Meier estimator. RESULTS IDO expression in tumor cells or APCs was not predictive for treatment response. The median OS was 26 months in IDO-positive and IDO-negative patients, regardless of IDO expression in tumor cells or APCs. A correlation of IHC expression scores of IDO and PD-L1 could not be documented. CONCLUSION The exact role of IDO in creating an immunosuppressive tumor environment and its reversal needs to be further elucidated.
Collapse
Affiliation(s)
| | - Christina Schmitt
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Simone M. Goldinger
- Department of Dermatology, University Hospital of Zurich, Zürich, Switzerland
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Katharina C. Kähler
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christian Hafner
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Nora Kramer
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Waltraud Fröhlich
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zürich, Switzerland
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Stefan Schliep
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Michael C. Kirchberger
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|
33
|
Esposito T, Pentimalli F, Giordano A, Cortellino S. Vitamins and dietary supplements in cancer treatment: is there a need for increased usage? Expert Rev Anticancer Ther 2025:1-24. [PMID: 40322898 DOI: 10.1080/14737140.2025.2501077] [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: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Vitamins are essential for homeostasis and proper functioning of organisms. These micronutrients prevent tumor onset by functioning as antioxidants and enzymatic cofactors involved in anti-stress and immune responses, modulating epigenetic regulators, and shaping the microbiota composition. Unbalanced diets and sedentary lifestyles contribute to obesity, associated with increasing cancer risk. Cancer patients often exhibit vitamin deficiencies due to chronic inflammation, anticancer therapies, and tumor-induced metabolic changes, leading to malnutrition and cachexia. AREAS COVERED This review critically analyzes preclinical and clinical studies, sourced from PubMed and ClinicalTrials.gov databases, that investigate the potential benefits of vitamin supplementation and dietary interventions, such as intermittent fasting and ketogenic diets, in mouse tumor models and cancer patients. This analysis elucidates the limitations of such interventions and suggests optimal dietary strategies to prevent cancer and enhance patients' quality of life and prognosis. EXPERT OPINION To date, clinical studies have found no substantial benefit of over-the-counter vitamin supplements and dietary interventions on cancer patients' health and prognosis. To prevent the spread of useless and potentially harmful products by the nutraceutical industry, establishing a regulatory authority is necessary to monitor and ensure product quality and validity before commercialization.
Collapse
Affiliation(s)
- Teresa Esposito
- Department of Clinical Dietetics and Metabolic Diseases, Cavalier Raffaele Apicella Hospital, ASL Napoli 3 Sud, Naples, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro", Bari, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Salvatore Cortellino
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, Italy
- S.H.R.O. Italia Foundation ETS, Turin, Italy
| |
Collapse
|
34
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response. Cell Rep 2025; 44:115698. [PMID: 40343794 DOI: 10.1016/j.celrep.2025.115698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/03/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy is experimentally challenging. Our approach, utilizing the Collaborative Cross mouse genetic resource, fixes the tumor genomic configuration while varying host genetics. We find that response to anti-PD-1 (aPD1) immunotherapy is significantly heritable in four distinct murine tumor models (H2: 0.18-0.40). For the MC38 colorectal carcinoma system, we map four significant ICI response quantitative trait loci (QTLs) with significant epistatic interactions. The differentially expressed genes within these QTLs that define responder genetics are highly enriched for processes involving antigen processing and presentation, allograft rejection, and graft vs. host disease (all p < 1 × 10-10). Functional blockade of two top candidate immune targets, GM-CSF and IL-2RB, completely abrogates the MC38 transcriptional response to aPD1 therapy. Thus, our in vivo experimental platform is a powerful approach for discovery of host genetic factors that establish the tumor immune microenvironment propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - John P Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | | | - Timothy M Stearns
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Jessica D S Grassmann
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - William F Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Elise T Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; OB/Gyn Department, UConn Health, Farmington, CT 06032, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - David V Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Laura G Reinholdt
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.
| |
Collapse
|
35
|
Zhu Y, Liu Q, Alffenaar JW, Wang S, Cao J, Dong S, Zhou X, Li X, Li X, Xiong H, Zhu L, Hu Y, Wang W. Gut Microbiota in Patients with Tuberculosis Associated with Different Drug Exposures of Antituberculosis Drugs. Clin Pharmacol Ther 2025. [PMID: 40326511 DOI: 10.1002/cpt.3687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025]
Abstract
Interindividual variability in drug exposure can significantly influence treatment outcomes and may lead to drug concentration-related side effects during tuberculosis (TB) treatment. Although the gut microbiota is known to affect drug metabolism, its impact on anti-TB drugs has not been thoroughly explored. This study sought to elucidate the relationship between pre-treatment gut microbiota and drug exposure levels among patients with pulmonary TB. Two cohorts were analyzed: a discovery cohort (N = 99) and a validation cohort (N = 32), both comprising patients undergoing anti-TB therapy with rifampicin, isoniazid, pyrazinamide, and ethambutol. The gut microbiota patterns of participants from the discovery cohort and the validation cohort were profiled by 16S rRNA gene sequencing and metagenomics, respectively. Analyses of both cohorts robustly established a positive association between pre-treatment microbial diversity and drug exposure, as well as significant differences in gut microbiota composition across various drug exposure groups. At the species level, Faecalibacterium prausnitzii was positively associated with drug exposure to rifampicin. Moreover, functional analysis revealed that starch and sucrose metabolism and secondary bile acid biosynthesis were more abundant in the high drug exposure group. To identify biomarkers capable of stratifying patients based on their drug exposure levels, 11 taxa, represented by Faecalibacterium, were selected in the discovery cohort (AUC = 0.992) and were confirmed in the validation cohort with high predictive accuracy (AUC = 0.894). This study demonstrated a correlation between microbial dysbiosis and reduced exposure to anti-TB medications. Optimizing treatment by regulating gut microbiota to improve drug exposure levels requires further validation through larger scale multicenter clinical trials.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Qiao Liu
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jan-Willem Alffenaar
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
- Department of Clinical Pharmacology, Westmead Hospital, Sydney, New South Wales, Australia
- Sydney Institute for Infectious Diseases, University of Sydney, Sydney, New South Wales, Australia
| | - Shanshan Wang
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Jiayi Cao
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Shulan Dong
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Xiangkang Zhou
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Xiaoxue Li
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Xuliang Li
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Haiyan Xiong
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Limei Zhu
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Yi Hu
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Laaraj J, Lachance G, Bergeron A, Fradet Y, Robitaille K, Fradet V. New insights into gut microbiota-prostate cancer crosstalk. Trends Mol Med 2025:S1471-4914(25)00087-5. [PMID: 40374457 DOI: 10.1016/j.molmed.2025.03.015] [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: 11/04/2024] [Revised: 01/24/2025] [Accepted: 03/28/2025] [Indexed: 05/17/2025]
Abstract
Recent evidence underscores a reciprocal relationship between the gut microbiota and prostate cancer (PCa). Dysbiosis, often driven by Western dietary habits and antibiotic use, can heighten systemic inflammation and hinder antitumor immunity, thereby fostering PCa onset and progression. Conversely, certain gut microbes and their metabolites may protect against tumor growth by modulating immune and hormonal pathways that impact therapeutic responses, including androgen deprivation therapy (ADT). Emerging evidence links gut microbial shifts to PCa aggressiveness, potentially sustaining local androgen production and promoting resistance. In this review, we explore current understanding of the gut-PCa interplay, highlighting key knowledge gaps and the need for further research to clarify how targeting the microbiome might influence PCa outcomes.
Collapse
Affiliation(s)
- Jalal Laaraj
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada
| | - Gabriel Lachance
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada
| | - Alain Bergeron
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Yves Fradet
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Karine Robitaille
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada
| | - Vincent Fradet
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada.
| |
Collapse
|
37
|
Cao M, Deng Y, Hao Q, Yan H, Wang QL, Dong C, Wu J, He Y, Huang LB, Xia X, Gao Y, Chen HN, Zhang WH, Zhang YJ, Zhuo X, Dai L, Hu H, Peng Y, Zhang F, Liu Z, Huang W, Zhang H, Yang L, Shu Y, Zhang W, Zhang Y, Xu H. Single-cell transcriptomic analysis reveals gut microbiota-immunotherapy synergy through modulating tumor microenvironment. Signal Transduct Target Ther 2025; 10:140. [PMID: 40312419 PMCID: PMC12045981 DOI: 10.1038/s41392-025-02226-7] [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: 07/29/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/03/2025] Open
Abstract
The gut microbiota crucially regulates the efficacy of immune checkpoint inhibitor (ICI) based immunotherapy, but the underlying mechanisms remain unclear at the single-cell resolution. Using single-cell RNA sequencing and subsequent validations, we investigate gut microbiota-ICI synergy by profiling the tumor microenvironment (TME) and elucidating critical cellular interactions in mouse models. Our findings reveal that intact gut microbiota combined with ICIs may synergistically increase the proportions of CD8+, CD4+, and γδ T cells, reduce glycolysis metabolism, and reverse exhausted CD8+ T cells into memory/effector CD8+ T cells, enhancing antitumor response. This synergistic effect also induces macrophage reprogramming from M2 protumor Spp1+ tumor-associated macrophages (TAMs) to Cd74+ TAMs, which act as antigen-presenting cells (APCs). These macrophage subtypes show a negative correlation within tumors, particularly during fecal microbiota transplantation. Depleting Spp1+ TAMs in Spp1 conditional knockout mice boosts ICI efficacy and T cell infiltration, regardless of gut microbiota status, suggesting a potential upstream role of the gut microbiota and highlighting the crucial negative impact of Spp1+ TAMs during macrophage reprogramming on immunotherapy outcomes. Mechanistically, we propose a γδ T cell-APC-CD8+ T cell axis, where gut microbiota and ICIs enhance Cd40lg expression on γδ T cells, activating Cd40 overexpressing APCs (e.g., Cd74+ TAMs) through CD40-CD40L-related NF-κB signaling and boosting CD8+ T cell responses via CD86-CD28 interactions. These findings highlight the potential importance of γδ T cells and SPP1-related macrophage reprogramming in activating CD8+ T cells, as well as the synergistic effect of gut microbiota and ICIs in immunotherapy through modulating the TME.
Collapse
Affiliation(s)
- Minyuan Cao
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huayun Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Quan-Lin Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Chunyan Dong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yajiao He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li-Bin Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuyang Xia
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaozhen Zhuo
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Huiyuan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yan Zhang
- Lung Cancer Center/Lung Cancer Institute, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China.
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
38
|
Wang J, Cheng W, Yang R. Nervous system-gut microbiota-immune system axis: future directions for preventing tumor. Front Immunol 2025; 16:1535955. [PMID: 40376000 PMCID: PMC12078214 DOI: 10.3389/fimmu.2025.1535955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025] Open
Abstract
Tumor is one of the leading causes of death worldwide. The occurrence and development of tumors are related to multiple systems and factors such as the immune system, gut microbiota, and nervous system. The immune system plays a critical role in tumor development. Studies have also found that the gut microbiota can directly or indirectly affect tumorigenesis and tumor development. With increasing attention on the tumor microenvironment in recent years, the nervous system has emerged as a novel regulator of tumor development. Some tumor therapies based on the nervous system have also been tested in clinical trials. However, the nervous system can not only directly interact with tumor cells but also indirectly affect tumor development through the gut microbiota. The nervous system-mediated gut microbiota can regulate tumorigenesis, growth, invasion, and metastasis through the immune system. Here, we mainly explore the potential effects of the nervous system-gut microbiota-immune system axis on tumorigenesis and tumor development. The effects of the nervous system-gut microbiota-immune system axis on tumors involve the nervous system regulating immune cells through the gut microbiota, which can prevent tumor development. Meanwhile, the direct effects of the gut microbiota on tumors and the regulation of the immune system by the nervous system, which can affect tumor development, are also reviewed.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
39
|
Zhang L, Li MJ, Li XP, Yang B, Xiao T, Wang P, Zhang WD. Respiratory microbiota diversity as a predictive biomarker for the efficacy of PD‑1 blockades in patients with advanced non‑small cell lung cancer: A retrospective exploratory study. Oncol Lett 2025; 29:251. [PMID: 40201032 PMCID: PMC11977453 DOI: 10.3892/ol.2025.14997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Despite advancements in immunotherapy, particularly regarding programmed cell death protein 1 (PD-1)/programmed death-ligand 1 blockades, the clinical outcomes in non-small cell lung cancer (NSCLC) remain variable with limited predictive biomarkers currently available. The present study investigated respiratory microbiota diversity as a potential biomarker to predict the efficacy of PD-1 blockades in patients with advanced NSCLC. A retrospective analysis was conducted on 60 patients treated with PD-1 blockades from May 2019 to May 2023. Clinical data were collected and respiratory microbiota from deep induced sputum specimens were analyzed using 16S rRNA gene sequencing. An index of respiratory microbiota α diversity was applied and exploratory analysis was performed accordingly. The objective response rate (ORR) and disease control rate among the 60 patients receiving PD-1 blockades was 23.3% (95% CI, 13.4-36.0%) and 58.3% (95% CI, 44.9-70.9%), respectively. Analysis of prognostic data of patients with advanced NSCLC receiving PD-1 blockades monotherapy demonstrated a median progression-free survival of 3.4 months (95% CI, 2.54-4.26) and a median overall survival (OS) of 12.3 months (95% CI, 6.29-18.31). Patients were stratified into high and low α diversity groups based on the Shannon diversity index of respiratory microbiota. The ORR was increased in the high diversity group (26.7%) compared with that of the low diversity group (20.0%), although the difference was not statistically significant (P=0.542). Notably, the high diversity group demonstrated a longer median PFS (3.9 vs. 2.8 months; P=0.017) and median OS (16.8 vs. 6.8 months; P=0.016) compared with that of the low diversity group. These findings suggested that PD-1 blockades demonstrate promising therapeutic activity for patients with previously treated advanced NSCLC in clinical practice. Respiratory microbiota α diversity might serve as a potential biomarker to predict the efficacy of PD-1 blockades monotherapy in patients with advanced NSCLC in the future. Therefore, further prospective studies are warranted to validate these findings and to explore the underlying mechanisms by which respiratory microbiota might modulate the immune response to cancer therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Ming-Jiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Xiao-Ping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Ting Xiao
- Department of Pharmacy, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, P.R. China
| | - Ping Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300202, P.R. China
| | - Wei-Dong Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| |
Collapse
|
40
|
Huang M, Zhang Y, Chen Z, Yu X, Luo S, Peng X, Li X. Gut microbiota reshapes the TNBC immune microenvironment: Emerging immunotherapeutic strategies. Pharmacol Res 2025; 215:107726. [PMID: 40184763 DOI: 10.1016/j.phrs.2025.107726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited treatment options and poor prognosis. The gut microbiota, a diverse community of microorganisms in the gastrointestinal tract, plays a crucial role in regulating immune responses through the gut-immune axis. Recent studies have highlighted its significant impact on TNBC progression and the efficacy of immunotherapies. This review examines the interactions between gut microbiota and the immune system in TNBC, focusing on key immune cells and pathways involved in tumor immunity. It also explores microbiota modulation strategies, including probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation, as potential methods to enhance immunotherapeutic outcomes. Understanding these mechanisms offers promising avenues for improving treatment efficacy and patient prognosis in TNBC.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Yikai Zhang
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Zhaoji Chen
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Xin Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian 350011, China
| | - Shiping Luo
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian 350011, China.
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China; Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management, China.
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning 110122, China; Department of Physiologyand Pharmacology, Karolinska Institutet, Solna 171 65, Sweden.
| |
Collapse
|
41
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
42
|
Okazawa-Sakai M, Sakai SA, Hyodo I, Horasawa S, Sawada K, Fujisawa T, Yamamoto Y, Boku S, Hayasaki Y, Isobe M, Shintani D, Hasegawa K, Egawa-Takata T, Ito K, Ihira K, Watari H, Takehara K, Yagi H, Kato K, Chiyoda T, Harano K, Nakamura Y, Yamashita R, Yoshino T, Aoki D. Gut microbiome associated with PARP inhibitor efficacy in patients with ovarian cancer. J Gynecol Oncol 2025; 36:e38. [PMID: 39453391 PMCID: PMC12099047 DOI: 10.3802/jgo.2025.36.e38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVE To investigate an association between the gut microbiome and efficacy of poly(ADP-ribose) polymerase inhibitors (PARPi) in ovarian cancer. METHODS This study conducted fecal microbiome analysis (16S rRNA gene sequencing) and circulating tumor DNA (ctDNA) profiling for ovarian cancer patients who underwent PARPi maintenance therapy. Fecal and blood samples were collected at the baseline and the progressive disease (PD) or last follow-up. The relative abundance of gut microbes and progression-free survival (PFS) were analyzed using linear discriminant analysis of effect size and the Cox proportional hazard model according to BRCA1/2 mutation (BRCA1/2mut) status detected by ctDNA sequencing. RESULTS Baseline samples were available from 23 BRCA1/2mut-positive patients and 33 BRCA1/2mut-negative patients. The microbes enriched in the baseline samples with long PFS were Bifidobacterium, Roseburia, Dialister, Butyricicoccus, and Bilophila for BRCA1/2mut-positive patients and Phascolarctobacterium for BRCA1/2mut-negative patients. In multivariate analyses dividing patients by the median values of relative abundances, no bacteria were associated with PFS in BRCA1/2mut-positive patients, whereas high Phascolarctobacterium abundances (≥1.11%) was significantly associated with longer PFS in BRCA1/2mut-negative patients (median 14.0 vs. 5.9 months, hazard ratio=0.28; 95% confidence interval=0.11-0.69; p=0.014). In the last samples, the relative abundances of Phascolarctobacterium were significantly higher in patients without PD (n=5) than those with PD (n=15) (median 1.25% vs. 0.06%; p=0.016). CONCLUSION High fecal composition of Phascolarctobacterium was associated with prolonged PFS in patients with BRCA1/2mut-negative ovarian cancer receiving PARPi therapy. Our results would provide new insights for future research.
Collapse
Affiliation(s)
- Mika Okazawa-Sakai
- Department of Gynecologic Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
- Department of Cancer Genomic Medicine, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Shunsuke A Sakai
- Department of Integrated Biosciences, Graduate School of Frontier Science, University of Tokyo, Kashiwa, Japan
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Ichinosuke Hyodo
- Department of Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Satoshi Horasawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kentaro Sawada
- Department of Medical Oncology, Kushiro Rosai Hospital, Kushiro, Japan
| | - Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yasuko Yamamoto
- Department of Hereditary Tumors, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan
| | - Yoh Hayasaki
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masanori Isobe
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Daisuke Shintani
- Department of Gynecology Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kosei Hasegawa
- Department of Gynecology Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Tomomi Egawa-Takata
- Department of Obstetrics and Gynecology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Kimihiko Ito
- Department of Obstetrics and Gynecology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Kei Ihira
- Department of Obstetrics and Gynecology, Hokkaido University Hospital, Sapporo, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University Hospital, Sapporo, Japan
| | - Kazuhiro Takehara
- Department of Gynecologic Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Hiroshi Yagi
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenichi Harano
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, University of Tokyo, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
- International University of Health and Welfare Graduate School, Otawara, Japan.
| |
Collapse
|
43
|
Hou X, Zhai L, Fu L, Lu J, Guo P, Zhang Y, Zheng D, Ma G. Advances in Engineered Phages for Disease Treatment. SMALL METHODS 2025; 9:e2401611. [PMID: 39935185 DOI: 10.1002/smtd.202401611] [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: 09/29/2024] [Revised: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Phage therapy presents a promising solution for combating multidrug-resistant (MDR) bacterial infections and other bacteria-related diseases, attributed to their innate ability to target and lyse bacteria. Recent clinical successes, particularly in treating MDR-related respiratory and post-surgical infections, validated the therapeutic potential of phage therapy. However, the complex microenvironment within the human body poses significant challenges to phage activity and efficacy in vivo. To overcome these barriers, recent advances in phage engineering have aimed to enhance targeting accuracy, improve stability and survivability, and explore synergistic combinations with other therapeutic modalities. This review provides a comprehensive overview of phage therapy, emphasizing the application of engineered phages in antibacterial therapy, tumor therapy, and vaccine development. Furthermore, the review highlights the current challenges and future trends for advancing phage therapy toward broader clinical applications.
Collapse
Affiliation(s)
- Xiaolin Hou
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Lin Zhai
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Laiying Fu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junna Lu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Diwei Zheng
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
44
|
Charalambous H, Brown C, Vogazianos P, Katsaounou K, Nikolaou E, Stylianou I, Papageorgiou E, Vraxnos D, Aristodimou A, Chi J, Costeas P, Shammas C, Apidianakis Y, Antoniades A. Dysbiosis in the Gut Microbiome of Pembrolizumab-Treated Non-Small Lung Cancer Patients Compared to Healthy Controls Characterized Through Opportunistic Sampling. Thorac Cancer 2025; 16:e70075. [PMID: 40356191 PMCID: PMC12069221 DOI: 10.1111/1759-7714.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The gut microbiome influences the host immune system, cancer development and progression, as well as the response to immunotherapy during cancer treatment. Here, we analyse the composition of the gut bacteriome in metastatic Non-Small Cell Lung Cancer (NSCLC) patients receiving Pembrolizumab immunotherapy within a prospective maintenance trial through opportunistic sampling during treatment. METHODS The gut microbiome profiles of NSCLC patients were obtained from stool samples collected during Pembrolizumab treatment and analysed with 16S rRNA metagenomics sequencing. Patient profiles were compared to a group of healthy individuals of matching ethnic group, age, sex, BMI and comorbidities. RESULTS A significant decrease in the treated patients was observed in two prominent bacterial families of the phylum Firmicutes, Lachnospiraceae and Ruminoccocaceae, which comprised 31.6% and 21.8% of the bacteriome in the healthy group but only 10.9% and 14.2% in the treated patient group, respectively. Species within the Lachnospiraceae and Ruminococcaceae families are known to break down undigested carbohydrates generating short chain fatty acids (SCFA), such as butyrate, acetate and propionate as their major fermentation end-products, which have been implicated in modifying host immune responses. In addition, a significant increase of the Bacteroidacaeae family (Bacteroidetes phylum) was observed from 10.7% in the healthy group to 23.3% in the treated patient group. Moreover, and in agreement with previous studies, a decrease in the Firmicutes to Bacteroidetes ratio in the metastatic NSCLC Pembrolizumab-treated patients was observed. CONCLUSION The observed differences indicate dysbiosis and a compromised intestinal health status in the metastatic NSCLC Pembrolizumab-treated patients. This data could inform future studies of immunotherapy treatment responses and modulation of the gut microbiome to minimise dysbiosis prior or concurrent to treatment. TRIAL REGISTRATION SWIPE Trial (NCT02705820).
Collapse
MESH Headings
- Humans
- Gastrointestinal Microbiome/drug effects
- Dysbiosis/chemically induced
- Dysbiosis/microbiology
- Dysbiosis/pathology
- Male
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/microbiology
- Female
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/microbiology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Aged
- Prospective Studies
- Case-Control Studies
- Antineoplastic Agents, Immunological/therapeutic use
Collapse
Affiliation(s)
| | | | - Paris Vogazianos
- Stremble Ventures LtdLimassolCyprus
- European University CyprusEngomiCyprus
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ramesh A, Srinivasan D, Subbarayan R, Chauhan A, Krishnamoorthy L, Kumar J, Krishnan M, Shrestha R. Enhancing Colorectal Cancer Treatment: The Role of Bifidobacterium in Modulating Gut Immunity and Mitigating Capecitabine-Induced Toxicity. Mol Nutr Food Res 2025; 69:e70023. [PMID: 40109200 DOI: 10.1002/mnfr.70023] [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: 12/11/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related mortality globally and presents significant challenges in treatment and patient care. Capecitabine, a widely used prodrug of 5-fluorouracil (5-FU), offers targeted delivery with reduced systemic toxicity compared to traditional chemotherapies. However, capacitabine is associated with adverse effects, such as hand-foot syndrome, gastrointestinal issues, and mucositis. Emerging evidence suggests that probiotics, particularly Bifidobacterium, play a pivotal role in gut microbiota modulation, promoting anti-inflammatory cytokines and short-chain fatty acids, such as butyrate, which possess both intestinal protective and anti-cancer properties. In this review, we explored the potential of Bifidobacterium to improve chemotherapy outcomes by mitigating inflammation and enhancing mucosal immunity in CRC patients. Furthermore, we demonstrated in silico approaches, including molecular docking and protein-protein interaction analysis, for Bifidobacterium and Toll-like receptor 2 (TLR-2), a key mediator of intestinal immunity. Docking results revealed strong binding affinity, suggesting the activation of anti-inflammatory pathways. Notably, this interaction enhanced IL-10 production while reducing pro-inflammatory cytokines, such as IL-6 and TNF-α, fostering gut homeostasis and mitigating chronic inflammation, a key driver of CRC progression. Therefore, future research should focus on personalized probiotics and validating their synergy with chemotherapy and immunotherapy to improve CRC treatment outcomes.
Collapse
Affiliation(s)
- Aswathi Ramesh
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Loganathan Krishnamoorthy
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Jeevan Kumar
- Department of Biomedical Sciences, The Apollo University, Chittoor, Andhra Pradesh, India
| | - Madhan Krishnan
- Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rupendra Shrestha
- Department of Natural and Applied Sciences, Nexus Institute of Research and Innovation (NIRI), Lalitpur, Nepal
| |
Collapse
|
46
|
Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025; 25:353-369. [PMID: 39748116 DOI: 10.1038/s41577-024-01111-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Cancers can avoid immune-mediated elimination by acquiring traits that disrupt antitumour immunity. These mechanisms of immune evasion are selected and reinforced during tumour evolution under immune pressure. Some immunogenic subclones are effectively eliminated by antitumour T cell responses (a process known as immunoediting), which results in a clonally selected tumour. Other cancer cells arise to resist immunoediting, which leads to a tumour that includes several distinct cancer cell populations (referred to as intratumour heterogeneity (ITH)). Tumours with high ITH are associated with poor patient outcomes and a lack of responsiveness to immune checkpoint blockade therapy. In this Review, we discuss the different ways that cancer cells evade the immune system and how these mechanisms impact immunoediting and tumour evolution. We also describe how subclonal antigen presentation in tumours with high ITH can result in immune evasion.
Collapse
Affiliation(s)
- Malte Roerden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Ragon Institute of Mass General Hospital, Massachusetts Institute for Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
47
|
Nobels A, van Marcke C, Jordan BF, Van Hul M, Cani PD. The gut microbiome and cancer: from tumorigenesis to therapy. Nat Metab 2025; 7:895-917. [PMID: 40329009 DOI: 10.1038/s42255-025-01287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 03/20/2025] [Indexed: 05/08/2025]
Abstract
The gut microbiome has a crucial role in cancer development and therapy through its interactions with the immune system and tumour microenvironment. Although evidence links gut microbiota composition to cancer progression, its precise role in modulating treatment responses remains unclear. In this Review, we summarize current knowledge on the gut microbiome's involvement in cancer, covering its role in tumour initiation and progression, interactions with chemotherapy, radiotherapy and targeted therapies, and its influence on cancer immunotherapy. We discuss the impact of microbial metabolites on immune responses, the relationship between specific bacterial species and treatment outcomes, and potential microbiota-based therapeutic strategies, including dietary interventions, probiotics and faecal microbiota transplantation. Understanding these complex microbiota-immune interactions is critical for optimizing cancer therapies. Future research should focus on defining microbial signatures associated with treatment success and developing targeted microbiome modulation strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Amandine Nobels
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- UCLouvain, Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
| | - Cédric van Marcke
- UCLouvain, Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bénédicte F Jordan
- UCLouvain, Université catholique de Louvain, Biomedical Magnetic Resonance group (REMA), Louvain Drug Research Institute (LDRI), Brussels, Belgium
| | - Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium.
| |
Collapse
|
48
|
Wu H, Liu J, Zhang XH, Jin S, Li P, Liu H, Zhao L, Wang J, Zhao S, Tian HD, Lai JR, Hao Y, Liu GR, Hou K, Yan M, Liu SL, Pang D. The combination of flaxseed lignans and PD-1/ PD-L1 inhibitor inhibits breast cancer growth via modulating gut microbiome and host immunity. Drug Resist Updat 2025; 80:101222. [PMID: 40048957 DOI: 10.1016/j.drup.2025.101222] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Patients with breast cancer (BC) who benefit from the PD-1/PD-L1 inhibitor (PDi) is limited, necessitating novel strategies to improve immunotherapy efficacy of BC. Here we aimed to investigate the inhibitory effects of flaxseed lignans (FL) on the biological behaviors of BC and evaluate the roles of FL in enhancing the anticancer effects of PDi. METHODS HPLC was used to detect the content of enterolactone (ENL), the bacterial transformation product of FL. Transcript sequencing was performed and identified CD38 as a downstream target gene of ENL. CD38-overexpressing cells were constructed and cell proliferation, colony formation, wound healing and transwell assays were used to assess the function of ENL/CD38 axis on BC cells in vitro. Multiplexed immunohistochemistry (mIHC) and CyTOF were used to detect the changes of the tumor immune microenvironment (TIM). 16S rDNA sequencing was used to explore the changes of gut microbiota in mice. A series of in vivo experiments were conducted to investigate the anticancer effects and mechanisms of FL and PDi. RESULTS FL was converted to ENL by gut microbiota and FL administration inhibited the progression of BC. ENL inhibited the malignant behaviors of BC by downregulating CD38, a key gene associated with immunosuppression and PD-1/PD-L1 blockade resistance. The mIHC assay revealed that FL administration enhanced CD3+, CD4+ and CD8+ cells and reduced F4/80+ cells in TIM. CyTOF confirmed the regulatory effects of FL and FL in combination with PDi (FLcPDi) on TIM. In addition, 16S rDNA analysis demonstrated that FLcPDi treatment significantly elevated the abundance of Akkermansia and, importantly, Akkermansia administration enhanced the response to PDi in mice treated with antibiotics. CONCLUSIONS The FL/ENL/CD38 axis inhibited BC progression. FL enhanced the anticancer effects of PDi by modulating gut microbiota and host immunity.
Collapse
Affiliation(s)
- Hao Wu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jiena Liu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xing-Hua Zhang
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Shengye Jin
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Li
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Huidi Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Liuying Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianyu Wang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shilu Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Da Tian
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Jin-Ru Lai
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China; Longhu People's Hospital, Shantou, China.
| | - Meisi Yan
- Department of Pathology, Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Da Pang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
49
|
Sawada K, Yamashita R, Sakai SA, Horasawa S, Yoshikawa A, Fujisawa T, Kadowaki S, Kato K, Ueno M, Oki E, Komatsu Y, Chiyoda T, Horita Y, Yasui H, Denda T, Satake H, Esaki T, Satoh T, Takahashi N, Yamazaki K, Matsuhashi N, Nishina T, Takeda H, Ohtsubo K, Ohta T, Tsuji A, Goto M, Kato T, Bando H, Tsuchihara K, Nakamura Y, Yoshino T. Microbiome Landscape and Association with Response to Immune Checkpoint Inhibitors in Advanced Solid Tumors: A SCRUM-Japan MONSTAR-SCREEN Study. CANCER RESEARCH COMMUNICATIONS 2025; 5:857-870. [PMID: 40341952 DOI: 10.1158/2767-9764.crc-24-0543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/04/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Although the gut microbiome is associated with cancer development and progression, little is known about the effects of the gut microbiome landscape and the efficacy of immune checkpoint inhibitors (ICI) across cancer types. We investigated the association between the microbiome, clinical features, and ICI efficacy across cancer types in a large nationwide screening project for solid tumors. Among 2,180 patients with advanced solid tumors enrolled in the SCRUM-Japan MONSTAR-SCREEN between October 2019 and September 2021, in the chemotherapy-naïve cohort (n = 817), a high prevalence of oral bacteria was observed in patients using proton pump inhibitors (PPI) and those with upper gastrointestinal cancers, particularly postoperative patients with gastric or pancreatic cancer. Among patients treated with ICIs (n = 333), a high abundance of sequence variants in the gut microbiome was not significantly associated with ICI efficacy across cancer types (HR = 0.94; 95% confidence interval, 0.73-1.21). However, high oral bacteria in feces significantly correlated with a shorter progression-free survival compared with low oral bacteria (median, 4.34 vs. 6.97 months; HR = 1.38; 95% confidence interval, 1.07-1.78). Notably, in patients using PPIs, a higher proportion of oral bacteria influenced progression-free survival outcomes of ICI treatment (median, 3.15 vs. 2.04 months; P = 0.08), unlike in PPI nonusers (median, 7.13 vs. 5.55 months; P = 0.74). This study of the gut microbiome has unveiled significant insights into its landscape and potential impact on ICI efficacy. It highlights that the abundance of oral bacteria in feces may play a critical role in diminishing ICI efficacy among patients using PPIs. SIGNIFICANCE As part of the MONSTAR-SCREEN, a prospective nationwide project for patients with solid tumors, we found that although gut microbiome diversity does not consistently predict ICI efficacy across cancer types, a high level of oral bacteria in the gut is linked to reduced ICI effectiveness, especially in patients using PPIs. These findings highlight the potential clinical impact of microbiome variations on cancer treatment outcomes.
Collapse
Affiliation(s)
- Kentaro Sawada
- Department of Medical Oncology, Kushiro Rosai Hospital, Kushiro, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Shunsuke A Sakai
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Satoshi Horasawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Ayumu Yoshikawa
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takao Fujisawa
- Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Ken Kato
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Eiji Oki
- Department of Surgery and Science, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yosuke Horita
- Department of Medical Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Tadamichi Denda
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Hironaga Satake
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Taroh Satoh
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Ina, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomohiro Nishina
- Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Gifu, Japan
| | - Hiroyuki Takeda
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Koushiro Ohtsubo
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Akihito Tsuji
- Department of Clinical Oncology, Kagawa University Hospital, Amagasaki, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
50
|
Andreu-Sánchez S, Blanco-Míguez A, Wang D, Golzato D, Manghi P, Heidrich V, Fackelmann G, Zhernakova DV, Kurilshikov A, Valles-Colomer M, Weersma RK, Zhernakova A, Fu J, Segata N. Global genetic diversity of human gut microbiome species is related to geographic location and host health. Cell 2025:S0092-8674(25)00416-7. [PMID: 40311618 DOI: 10.1016/j.cell.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/23/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
The human gut harbors thousands of microbial species, each exhibiting significant inter-individual genetic variability. Although many studies have associated microbial relative abundances with human-health-related phenotypes, the substantial intraspecies genetic variability of gut microbes has not yet been comprehensively considered, limiting the potential of linking such genetic traits with host conditions. Here, we analyzed 32,152 metagenomes from 94 microbiome studies across the globe to investigate the human microbiome intraspecies genetic diversity. We reconstructed 583 species-specific phylogenies and linked them to geographic information and species' horizontal transmissibility. We identified 484 microbial-strain-level associations with 241 host phenotypes, encompassing human anthropometric factors, biochemical measurements, diseases, and lifestyle. We observed a higher prevalence of a Ruminococcus gnavus clade in nonagenarians correlated with distinct plasma bile acid profiles and a melanoma and prostate-cancer-associated Collinsella clade. Our large-scale intraspecies genetic analysis highlights the relevance of strain diversity as it relates to human health.
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Daoming Wang
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Davide Golzato
- Department of CIBIO, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of CIBIO, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of CIBIO, University of Trento, Trento, Italy
| | | | - Daria V Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Mireia Valles-Colomer
- Department of CIBIO, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| | - Nicola Segata
- Department of CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|