1
|
He Y, Zhao G, Ouyang X, Wang S, Chen Y, Li C, He Y, Gao J, Han S, Zhao J, Wang J, Wang C. Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding. Gut Microbes 2025; 17:2489072. [PMID: 40205678 PMCID: PMC11988229 DOI: 10.1080/19490976.2025.2489072] [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: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.
Collapse
Affiliation(s)
- Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Ouyang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Lee AH, Rodriguez Jimenez DM, Meisel M. Limosilactobacillus reuteri - a probiotic gut commensal with contextual impact on immunity. Gut Microbes 2025; 17:2451088. [PMID: 39825615 DOI: 10.1080/19490976.2025.2451088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
The gut microbiome plays a key role in human health, influencing various biological processes and disease outcomes. The historical roots of probiotics are traced back to Nobel Laureate Élie Metchnikoff, who linked the longevity of Bulgarian villagers to their consumption of sour milk fermented by Lactobacilli. His pioneering work led to the global recognition of probiotics as beneficial supplements, now a multibillion-dollar industry. Modern probiotics have been extensively studied for their immunomodulatory effects. Limosilactobacillus reuteri (L. reuteri), a widely used probiotic, has garnered significant attention for its systemic immune-regulatory properties, particularly in relation to autoimmunity and cancer. This review delves into the role of L. reuteri in modulating immune responses, with a focus on its impact on systemic diseases.
Collapse
Affiliation(s)
- Amanda H Lee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Huang M, Ji Q, Huang H, Wang X, Wang L. Gut microbiota in hepatocellular carcinoma immunotherapy: immune microenvironment remodeling and gut microbiota modification. Gut Microbes 2025; 17:2486519. [PMID: 40166981 PMCID: PMC11970798 DOI: 10.1080/19490976.2025.2486519] [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: 10/02/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality, with limited treatment options at advanced stages. The gut microbiota, a diverse community of microorganisms residing in the gastrointestinal tract, plays a pivotal role in regulating immune responses through the gut-liver axis. Emerging evidence underscores its impact on HCC progression and the efficacy of immunotherapy. This review explores the intricate interactions between gut microbiota and the immune system in HCC, with a focus on key immune cells and pathways involved in tumor immunity. Additionally, it highlights strategies for modulating the gut microbiota - such as fecal microbiota transplantation, dietary interventions, and probiotics - as potential approaches to enhancing immunotherapy outcomes. A deeper understanding of these mechanisms could pave the way for novel therapeutic strategies aimed at improving patient prognosis.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian, China
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Quansong Ji
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huiyan Huang
- Ward 3, De’an Hospital, Xianyou County, Putian, Fujian, China
| | - Xiaoqian Wang
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Lin Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Plata-Gómez AB, Ho PC. Age- and diet-instructed metabolic rewiring of the tumor-immune microenvironment. J Exp Med 2025; 222:e20241102. [PMID: 40214641 PMCID: PMC11987706 DOI: 10.1084/jem.20241102] [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: 12/20/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The tumor-immune microenvironment (TIME) plays a critical role in tumor development and metastasis, as it influences the evolution of tumor cells and fosters an immunosuppressive state by intervening the metabolic reprogramming of infiltrating immune cells. Aging and diet significantly impact the metabolic reprogramming of the TIME, contributing to cancer progression and immune evasion. With aging, immune cell function declines, leading to a proinflammatory state and metabolic alterations such as increased oxidative stress and mitochondrial dysfunction, which compromise antitumor immunity. Similarly, dietary factors, particularly high-fat and high-sugar diets, promote metabolic shifts, creating a permissive TIME by fostering tumor-supportive immune cell phenotypes while impairing the tumoricidal activity of immune cells. In contrast, dietary restrictions have been shown to restore immune function by modulating metabolism and enhancing antitumor immune responses. Here, we discuss the intricate interplay between aging, diet, and metabolic reprogramming in shaping the TIME, with a particular focus on T cells, and highlight therapeutic strategies targeting these pathways to empower antitumor immunity.
Collapse
Affiliation(s)
- Ana Belén Plata-Gómez
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
8
|
Cheng MC, Chen HM, Chang TY, Chen MC, Bai BJ, Chao CH, Hsieh WY, Lin YT, Ni CK, Lu MK, Liu HK, Lee SS, Chang CC. Acetylated glucomanno-oligosaccharides from human gut microbial degradation of Dendrobium polysaccharides: production and effects on anti-hyperglycemia-related factors. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2025; 10:100808. [DOI: 10.1016/j.carpta.2025.100808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025] Open
|
9
|
Majenka P, Hoffmann M, Strobel S, Rötzer I, Enk A, Hassel JC. Influence of high-fiber diet on ipilimumab-induced gastrointestinal toxicity in metastatic melanoma. Clin Nutr ESPEN 2025; 67:660-664. [PMID: 40112923 DOI: 10.1016/j.clnesp.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND The anti-cytotoxic T-lymphocyte-associated protein (CTLA)-4 antibody ipilimumab (ipi) and the anti-programmed death (PD)-1 antibody nivolumab (nivo) are routinely used to treat metastatic melanoma. One of the most frequent severe immune-related adverse events (irAEs) induced by ipi is diarrhea as a symptom of ir-colitis. Here, the composition of the gut microbiome was shown to correlate with the risk of developing colitis. Stimulated by a patient case and the knowledge that nutrition influences the gut microbiome, we performed a retrospective analysis to evaluate dietary habits and the frequency of colitis in patients with ipi ± nivo therapy. METHODS Patients with metastasized stage III or IV melanoma who were treated with ipi ± nivo and who were willing to take part in a nutritional survey and interview at least three months after the first ipi dose were included into the study. Dietary habits were investigated using the food frequency questionnaire (FFQ) and personal interviews. The calculated daily intake of calories, carbohydrates, fats, proteins, sugars, and dietary fiber was correlated with the development of ir-colitis. RESULTS 20 patients were included into this study, and all but one received ipi-nivo combination therapy. The median age was 59.5 years, and 60 % were male. 4 of 20 patients (20 %) developed ir-colitis grade 3 after two cycles in the median and were managed with at least high-dose corticosteroids. The FFQ and interview were conducted in a median of six months after treatment initiation. In general, the interviewed patients followed a typical western-pattern diet based on carbohydrates as the main, followed by fat as the second most important energy substrate. Comparing patients with and without colitis our investigation revealed that the achieved amount of recommended dietary fiber intake per total energy intake (TEI) was negatively associated with diarrhea and colitis (p = 0.061). No significant differences concerning daily intake of calories, carbohydrates, fats, proteins, and sugar were found. In addition, no significant differences were found among patients in terms of their age, gender, tobacco use, supplement intake, therapy regime, or body mass index (BMI). CONCLUSIONS This pilot study gives first hints that nutritional habits might influence treatment tolerability to ipi ± nivo therapy. A high-fiber diet might protect against ir-colitis and diarrhea in ipi-treated patients. This observation should be validated by a prospective randomized interventional trial. However, if it is possible to prevent ir-colitis by a high-fiber diet that would be of great impact on routine patient treatment.
Collapse
Affiliation(s)
- Pawel Majenka
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Hoffmann
- Department of Nutrition Therapy, National Center for Tumor Diseases, Heidelberg, Germany
| | - Sophia Strobel
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Ingeborg Rötzer
- Department of Nutrition Therapy, National Center for Tumor Diseases, Heidelberg, Germany
| | - Alexander Enk
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Roichman A, Zuo Q, Hwang S, Lu W, Cordova RA, MacArthur MR, Boyer JA, Mitchell SJ, Powers J, Koval SA, Hunter CJ, Rijmers J, Ryseck RP, AbuSalim JE, Chatterjee S, Lee WD, Xu X, Xing X, Chen Z, Zeng X, Marwaha S, McBride MJ, Guo JY, Kang Y, Donia MS, Rabinowitz JD. Microbiome metabolism of dietary phytochemicals controls the anticancer activity of PI3K inhibitors. Cell 2025:S0092-8674(25)00511-2. [PMID: 40393457 DOI: 10.1016/j.cell.2025.04.041] [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: 08/26/2024] [Revised: 03/04/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Phosphatidylinositol 3-kinase (PI3K) signaling is both the effector pathway of insulin and among the most frequently activated pathways in human cancer. In murine cancer models, the efficacy of PI3K inhibitors is dramatically enhanced by a ketogenic diet, with a proposed mechanism involving dietary suppression of insulin. Here, we confirm profound diet-PI3K anticancer synergy but show that it is, surprisingly, unrelated to diet macronutrient composition. Instead, the diet-PI3K interaction involves microbiome metabolism of ingested phytochemicals. Specifically, murine ketogenic diet lacks the complex spectrum of phytochemicals found in standard chow, including the soy phytochemicals soyasaponins. We find that soyasaponins are converted by the microbiome into inducers of hepatic cytochrome P450 enzymes, and thereby lower PI3K inhibitor blood levels and anticancer activity. A high-carbohydrate, low-phytochemical diet synergizes with PI3K inhibition to treat cancer in mice, as do antibiotics that curtail the gut microbiome. Thus, diet impacts anticancer drug activity through phytochemical-microbiome-liver interactions.
Collapse
Affiliation(s)
- Asael Roichman
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Qianying Zuo
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sunghoon Hwang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Ricardo A Cordova
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Jacob A Boyer
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Sarah J Mitchell
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Jesse Powers
- Rutgers Cancer Institute, New Brunswick, NJ, USA
| | - Sophia A Koval
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Jamie Rijmers
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Rolf-Peter Ryseck
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Jenna E AbuSalim
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Seema Chatterjee
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Xincheng Xu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Xi Xing
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zihong Chen
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Xianfeng Zeng
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Siddharth Marwaha
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Matthew J McBride
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Jessie Y Guo
- Rutgers Cancer Institute, New Brunswick, NJ, USA
| | - Yibin Kang
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Nobels A, van Marcke C, Jordan BF, Van Hul M, Cani PD. The gut microbiome and cancer: from tumorigenesis to therapy. Nat Metab 2025:10.1038/s42255-025-01287-w. [PMID: 40329009 DOI: 10.1038/s42255-025-01287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
Moomin A, Gawusu S, Sidik AI, Tizumah MW, Adam MK, Adu-Gyamfi PKT, Mensah KB. Dietary Fiber and Cancer Management: A Twenty-Five-Year Bibliometric Review of Research Trends and Directions. BIOMED RESEARCH INTERNATIONAL 2025; 2025:5086946. [PMID: 40330094 PMCID: PMC12055321 DOI: 10.1155/bmri/5086946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025]
Abstract
Background: The global burden of cancer necessitates innovative approaches to its management and treatment. Traditional treatments like radiotherapy, immunotherapy, surgery, and chemotherapy, carry significant side effects that impact patient quality of life. Dietary fiber has attracted research interest as a potential mitigator of cancer progression and a supportive agent in cancer treatment. This bibliometric study analyzes trends in research connecting dietary fiber, cancer therapy, and gut health from April 1999 to May 2024. Methods and Results: Web of Science, PubMed, and Scopus databases were used to retrieve peer-reviewed publications from April 1999 to May 2024 on dietary fiber and cancer management. The study identifies a rising scholarly interest in dietary fiber's role in cancer management, focusing significantly on the interactions between dietary fiber and gut microbiota. These interactions are particularly noted for their influence on inflammation and cancer metastasis. The study highlights evolving research themes, the importance of specific fiber types in cancer progression, and highlights persistent foundational themes like glycosylation. Emerging areas include personalized nutrition and innovative therapeutic approaches. The geographical and institutional contributions, mainly from the United States and China, underline the significance of collaborative and interdisciplinary efforts in advancing research. Conclusion: This analysis emphasizes integrating dietary strategies in comprehensive cancer care and aims to address research gaps to develop more effective and patient-centered cancer therapy and prevention strategies.
Collapse
Affiliation(s)
- Aliu Moomin
- Rowett Institute, University of Aberdeen, Aberdeen, Scotland, UK
- Aberdeen Cancer Centre, University of Aberdeen, Aberdeen, Scotland, UK
| | - Sidique Gawusu
- Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Marizuk Waris Tizumah
- Department of Medical laboratory/Medical Imaging Technology, Accra Technical University, Accra, Greater Accra, Ghana
- Medical Centre, University of Ghana, Accra, Greater Accra, Ghana
| | - Maridia Kunateh Adam
- Department of Data Science, University of East London, London, England, UK
- Department of Dietetics, Robert Gordon University, Aberdeen, Scotland, UK
| | | | - Kwesi Boadu Mensah
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ashanti Region, Ghana
| |
Collapse
|
22
|
Adlakha YK, Chhabra R. The human microbiome: redefining cancer pathogenesis and therapy. Cancer Cell Int 2025; 25:165. [PMID: 40296128 PMCID: PMC12039184 DOI: 10.1186/s12935-025-03787-x] [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: 09/27/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
The human microbiome has always been an important determinant of health and recently, its role has also been described in cancer. The altered microbiome could aid cancer progression, modulate chemoresistance and significantly alter drug efficacy. The broad implications of microbes in cancer have prompted researchers to investigate the microbe-cancer axis and identify whether modifying the microbiome could sensitize cancer cells for therapy and improve the survival outcome of cancer patients. The preclinical data has shown that enhancing the number of specific microbial species could restore the patients' response to cancer drugs and the microbial biomarkers may play a vital role in cancer diagnostics. The elucidation of detailed interactions of the human microbiota with cancer would not only help identify the novel drug targets but would also enhance the efficacy of existing drugs. The field exploring the emerging roles of microbiome in cancer is at a nascent stage and an in-depth scientific perspective on this topic would make it more accessible to a wider audience. In this review, we discuss the scientific evidence connecting the human microbiome to the origin and progression of cancer. We also discuss the potential mechanisms by which microbiota affects initiation of cancer, metastasis and chemoresistance. We highlight the significance of the microbiome in therapeutic outcome and evaluate the potential of microbe-based cancer therapy.
Collapse
Affiliation(s)
- Yogita K Adlakha
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Sector-125, Noida, Uttar Pradesh, 201303, India.
| | - Ravindresh Chhabra
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
23
|
Verdys P, Johansen AZ, Gupta A, Presti M, Dionisio E, Madsen DH, Curioni-Fontecedro A, Donia M. Acquired resistance to immunotherapy in solid tumors. Trends Mol Med 2025:S1471-4914(25)00061-9. [PMID: 40274520 DOI: 10.1016/j.molmed.2025.03.010] [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: 11/28/2024] [Revised: 02/11/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025]
Abstract
Acquired resistance to immunotherapy (ARI) is a major challenge in solid tumors, limiting long-term success in up to 65% of patients who initially respond to immunotherapy. Defining ARI clinically remains complex, but ongoing efforts aim to establish standardized criteria. This review describes recent insights into ARI, revealing complex mechanisms involving both tumor-intrinsic mechanisms - such as antigen loss and presentation defects, interferon γ (IFNγ) insensitivity, tumor-mediated T cell exclusion, and metabolic reprogramming - as well as extrinsic factors such as inhibitory molecule upregulation, immunosuppressive cells, extracellular matrix (ECM) remodeling, and dysbiotic microbiota. Understanding the development of ARI is crucial for prevention and effective interventions. The integration of innovative strategies and translational research on appropriately collected samples is key to overcoming ARI and ensuring durable benefits for patients.
Collapse
Affiliation(s)
- Perrine Verdys
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Astrid Z Johansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Anurag Gupta
- Department of Medical Oncology, University of Fribourg, Fribourg, Switzerland
| | - Mario Presti
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Edoardo Dionisio
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Daniel H Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| |
Collapse
|
24
|
Du J, Guan Y, Zhang E. Regulatory role of gut microbiota in immunotherapy of hepatocellular carcinoma. Hepatol Int 2025:10.1007/s12072-025-10822-6. [PMID: 40229514 DOI: 10.1007/s12072-025-10822-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The gut microbiota plays a role in triggering innate immunity and regulating the immune microenvironment (IME) of hepatocellular carcinoma (HCC) by acting on various signaling receptors and transcription factors through its metabolites and related molecules. Furthermore, there is an increasing recognition of the gut microbiota as a potential therapeutic target for HCC, given its ability to modulate the efficacy of immune checkpoint inhibitors (ICIs). OBJECTIVE This review will discuss the mechanisms of gut microbiota in modulating immunotherapy of HCC, the predictive value of efficacy, and the therapeutic strategies for modulating the gut microbiota in detail. METHODS We conducted a systematic literature search in PubMed, Embase, Scopus, Cochrane Library, China National Knowledge Infrastructure, and Wanfang Chinese databases for articles involving the influence of gut microbiota on HCC immunotherapy. RESULTS The mechanisms underlying the effect of gut microbiota on HCC immunotherapy include gut-liver axis, tumor immune microenvironment (TIME), and antibodies. Patients who benefit from ICIs exhibit a higher abundance of gut microbiota. Antibiotics, fecal microbiota transplantation (FMT), probiotics, and prebiotics are effective methods to regulate gut microbiota. CONCLUSION The strong connection between the liver and gut will provide numerous opportunities for the development of microbiome-based diagnostics, treatments, or prevention strategies for HCC patients.
Collapse
Affiliation(s)
- Jiajia Du
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Yan Guan
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Erlei Zhang
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| |
Collapse
|
25
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [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] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
26
|
Liu S, Liu J, Mei Y, Zhang W. Gut microbiota affects PD-L1 therapy and its mechanism in melanoma. Cancer Immunol Immunother 2025; 74:169. [PMID: 40214675 PMCID: PMC11992302 DOI: 10.1007/s00262-025-04018-y] [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: 01/16/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 blockade, have shown great success in treating melanoma. PD-L1 (B7-H1, CD274), a ligand of PD-1, binds to PD-1 on T cells, inhibiting their activation and proliferation through multiple pathways, thus dampening tumor-reactive T cell activity. Studies have linked PD-L1 expression in melanoma with tumor growth, invasion, and metastasis, making the PD-1/PD-L1 pathway a critical target in melanoma therapy. However, immune-related adverse events are common, reducing the effectiveness of anti-PD-L1 treatments. Recent evidence suggests that the gut microbiome significantly influences anti-tumor immunity, with the microbiome potentially reprogramming the tumor microenvironment and overcoming resistance to anti-PD-1 therapies in melanoma patients. This review explores the mechanisms of PD-1/PD-L1 in melanoma and examines how gut microbiota and its metabolites may help address resistance to anti-PD-1 therapy, offering new insights for improving melanoma treatment strategies.
Collapse
Affiliation(s)
- Shiqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jiahui Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yingwu Mei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Wenjuan Zhang
- Beijing Life Science Academy (BLSA), Beijing, China.
- Key Laboratory of Tobacco Flavor Basic Research of CNTC, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, China.
| |
Collapse
|
27
|
Luo Z, Chen S, Chen P, Xiong K, Cao C. Association of dietary inflammatory index, composite dietary antioxidant index and risk of death among adult cancer survivors: findings from the National Health and Nutrition Examination Survey 2001-2018. Front Immunol 2025; 16:1556828. [PMID: 40260237 PMCID: PMC12009815 DOI: 10.3389/fimmu.2025.1556828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Background The development and progression of cancer can be impacted by the nutrients and components contained in the diet. This research seeks to explore the relationship between the antioxidant and pro-inflammatory properties of diet and the risk of all-cause mortality among cancer survivors. Methods Adults aged 20 and above who had been diagnosed with cancer and participated in the National Health and Nutrition Examination Survey (NHANES) from 2001 to 2018 were selected for this study. Their survival status was verified using death certificate information from the National Death Index. The study employed two established measures, the Composite Dietary Antioxidant Index (CDAI) and the Dietary Inflammatory Index (DII), to evaluate the antioxidant and inflammatory properties of participants' diets. A non-linear association between these two dietary indices and mortality was examined respectively using restricted cubic spline (RCS) regression. To quantify the relationship between the indices and mortality risk, multivariable Cox proportional hazards models were employed, generating hazard ratios and corresponding 95% confidence intervals. Furthermore, the study also explored the connection between the CDAI and DII. Results In this study, a total of 3,507 cancer survivors, representing an estimated 20,016,255 cancer survivors in the US, were included in the baseline analysis. The results showed that patients with lower DII or higher CDAI values had better survival rates. RCS regression revealed that both indicators showed linear relationships with all-cause mortality in the crude and adjusted models. It was consistently noted higher CDAI or lower DII was related to a reduced risk of all-cause mortality in cancer survivors in the Cox regression. Moreover, the subgroup analysis demonstrated that these associations hold true across various subgroups, lending credibility to the overall findings of the study. At last, an inverse correlation was observed between CDAI and DII in the diets of cancer survivors. Conclusion The research suggests that adopting a diet that low in pro-inflammatory foods and high in antioxidants may lower the all-cause mortality in cancer survivors. However, further prospective cohort studies are necessary to confirm these findings.
Collapse
Affiliation(s)
| | | | | | | | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
28
|
Roichman A, Reyes-Castellanos G, Chen Z, Chen Z, Mitchell SJ, MacArthur MR, Sawant A, Levett L, Powers J, Gomez M, Ibrahim M, Xu X, Tomlinson B, Hang X, Wei Y, Kang Y, White E, Rabinowitz JD. Lack of consistent effect of dietary fiber on immune checkpoint blockade efficacy across diverse murine tumor models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645975. [PMID: 40235983 PMCID: PMC11996298 DOI: 10.1101/2025.03.28.645975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Immune checkpoint blockade (ICB) has transformed cancer treatment, but success rates remain low in most cancers. Recent research suggest that dietary fiber enhances ICB response in melanoma patients and murine preclinical models through microbiome-dependent mechanisms. Yet, the robustness of this effect across cancer types and dietary contexts remains unclear. Specifically, prior literature compared grain-based chow (high fiber) to low-fiber purified diet, but these diets differ also on other dimensions including phytochemicals. Here we investigated, in mice fed grain-based chow or purified diets with differing quantities of isolated fibers (cellulose and inulin), metabolite levels and ICB activity in multiple tumor models. The blood and fecal metabolome were relatively similar between mice fed high- and low-fiber purified diets, but differed massively between mice fed purified diets or chow, identifying the factor as diet type, independent of fiber. Tumor growth studies in three implantable and two spontaneous genetically engineered tumor models revealed that fiber has a weaker impact on ICB (anti-PD-1) efficacy than previously reported. In some models, dietary modulation impacted ICB activity, but not in a consistent direction across models. In none of the models did we observe the pattern expected if fiber controlled ICB efficacy: strong efficacy in both chow and high-fiber purified diet but low efficacy in low-fiber purified diet. Thus, dietary fiber appears to have limited or inconsistent effect on ICB efficacy in mouse models, and other dietary factors that correlate with fiber intake may underlie the clinical correlations between fiber consumption and immunotherapy outcomes.
Collapse
|
29
|
Clavijo-Salomon MA, Trinchieri G. Unlocking the power of the microbiome for successful cancer immunotherapy. J Immunother Cancer 2025; 13:e011281. [PMID: 40180421 PMCID: PMC11966956 DOI: 10.1136/jitc-2024-011281] [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: 12/12/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
In recent years, evidence has shown that the gut microbiome significantly influences responses to immunotherapy. This has sparked interest in targeting it to improve therapy outcomes and predictions of response and toxicity. Research has demonstrated that dysbiosis, often resulting from antibiotic use, can diminish the effectiveness of immune checkpoint inhibitors, and this lack of efficacy could be linked to systemic inflammation. Certain bacterial species have been identified as having beneficial and harmful effects on immunotherapy in the clinic. While a clear consensus has yet to emerge on the optimal species for therapeutic use, introducing a new microbiome into immunotherapy-refractory patients may boost their chances of responding to further treatment attempts. State-of-the-art interventions targeting the microbiome-such as fecal microbiota transplantation-are being assessed clinically for their safety and potential to enhance treatment outcomes, with promising results. Additionally, the microbiome has been leveraged for its power to predict clinical outcomes using machine learning, and surprisingly, its predictive capability is comparable to that of other described multi-biomarker clinical scores. Here, we discuss developing knowledge concerning the microbiome's significance in cancer immunotherapy and outline future strategies for maximizing its potential in immuno-oncology.
Collapse
Affiliation(s)
- Maria A Clavijo-Salomon
- Laboratory of Integrative Cancer Immunology, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Gilbert JA, Azad MB, Bäckhed F, Blaser MJ, Byndloss M, Chiu CY, Chu H, Dugas LR, Elinav E, Gibbons SM, Gilbert KE, Henn MR, Ishaq SL, Ley RE, Lynch SV, Segal E, Spector TD, Strandwitz P, Suez J, Tropini C, Whiteson K, Knight R. Clinical translation of microbiome research. Nat Med 2025; 31:1099-1113. [PMID: 40217076 DOI: 10.1038/s41591-025-03615-9] [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: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
The landscape of clinical microbiome research has dramatically evolved over the past decade. By leveraging in vivo and in vitro experimentation, multiomic approaches and computational biology, we have uncovered mechanisms of action and microbial metrics of association and identified effective ways to modify the microbiome in many diseases and treatment modalities. This Review explores recent advances in the clinical application of microbiome research over the past 5 years, while acknowledging existing barriers and highlighting opportunities. We focus on the translation of microbiome research into clinical practice, spearheaded by Food and Drug Administration (FDA)-approved microbiome therapies for recurrent Clostridioides difficile infections and the emerging fields of microbiome-based diagnostics and therapeutics. We highlight key examples of studies demonstrating how microbiome mechanisms, metrics and modifiers can advance clinical practice. We also discuss forward-looking perspectives on key challenges and opportunities toward integrating microbiome data into routine clinical practice, precision medicine and personalized healthcare and nutrition.
Collapse
Affiliation(s)
- Jack A Gilbert
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin J Blaser
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Mariana Byndloss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Fransisco, San Francisco, CA, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Fransisco, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Hiutung Chu
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines, La Jolla, CA, USA
| | - Lara R Dugas
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Microbiome and Cancer Division, DKFZ, Heidelberg, Germany
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Katharine E Gilbert
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | | | - Suzanne L Ishaq
- School of Food and Agriculture, University of Maine, Orono, ME, USA
- Microbes and Social Equity working group, Orono, ME, USA
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - Susan V Lynch
- Benioff Center for Microbiome Medicine, Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- ZOE Ltd, London, UK
| | | | - Jotham Suez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carolina Tropini
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
31
|
Brandlmaier M, Koelblinger P. The discrepancy between managing the disease and a disease management program in systemic melanoma therapy. J Dtsch Dermatol Ges 2025; 23:527-531. [PMID: 40066483 PMCID: PMC11979555 DOI: 10.1111/ddg.15645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/26/2024] [Indexed: 04/10/2025]
Affiliation(s)
- Matthias Brandlmaier
- Department of Dermatology and AllergologyParacelsus Medical University Hospital Salzburg, Austria
| | - Peter Koelblinger
- Department of Dermatology and AllergologyParacelsus Medical University Hospital Salzburg, Austria
| |
Collapse
|
32
|
Delzenne NM, Bindels LB, Neyrinck AM, Walter J. The gut microbiome and dietary fibres: implications in obesity, cardiometabolic diseases and cancer. Nat Rev Microbiol 2025; 23:225-238. [PMID: 39390291 DOI: 10.1038/s41579-024-01108-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/12/2024]
Abstract
Dietary fibres constitute a heterogeneous class of nutrients that are key in the prevention of various chronic diseases. Most dietary fibres are fermented by the gut microbiome and may, thereby, modulate the gut microbial ecology and metabolism, impacting human health. Dietary fibres may influence the occurrence of specific bacterial taxa, with this effect varying between individuals. The effect of dietary fibres on microbial diversity is a matter of debate. Most intervention studies with dietary fibres in the context of obesity and related metabolic disorders reveal the need for an accurate assessment of the microbiome to better understand the variable response to dietary fibres. Epidemiological studies confirm that a high dietary fibre intake is strongly associated with a reduced occurrence of many types of cancer. However, there is a need to determine the impact of intervention with specific dietary fibres on cancer risk, therapy efficacy and toxicity, as well as in cancer cachexia. In this Review, we summarize the mechanisms by which the gut microbiome can mediate the physiological benefits of dietary fibres in the contexts of obesity, cardiometabolic diseases and cancer, their incidence being clearly linked to low dietary fibre intake.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
33
|
Golshani M, Taylor JA, Woolbright BL. Understanding the microbiome as a mediator of bladder cancer progression and therapeutic response. Urol Oncol 2025; 43:254-265. [PMID: 39117491 DOI: 10.1016/j.urolonc.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/17/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Bladder cancer (BCa) remains a significant source of morbidity and mortality. BCa is one of the most expensive tumors to treat, in part because of a lack of nonsurgical options. The recent advent of immunotherapy, alone or in combination with other compounds, has improved therapeutic options. Resistance to immunotherapy remains common, and many patients do not have durable response. Recent advances indicate immunotherapy efficacy may be tied in part to the endogenous bacteria present in our body, more commonly referred to as the microbiome. Laboratory and clinical data now support the idea that a healthy microbiome is critical to effective response to immunotherapy. At the same time, pathogenic interactions between the microbiome and immune cells can also serve to drive formation of tumors, increasing the complexity of these interactions. Given the rising importance of immunotherapy in BCa, understanding how we might be able to alter the microbiome to improve therapeutic efficacy offers a novel route to improved patient care. The goal of this review is to examine our current understanding of microbial interactions with the immune system and cancer with an emphasis on BCa. We will further attempt to define both current gaps in knowledge and future directions that may yield beneficial results to the field.
Collapse
Affiliation(s)
- Mahgol Golshani
- School of Medicine, University of Kansas Medical Center, Kansas City, KS
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS; Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | | |
Collapse
|
34
|
Zitvogel L, Derosa L, Routy B, Loibl S, Heinzerling L, de Vries IJM, Engstrand L, Segata N, Kroemer G. Impact of the ONCOBIOME network in cancer microbiome research. Nat Med 2025; 31:1085-1098. [PMID: 40217075 DOI: 10.1038/s41591-025-03608-8] [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: 08/02/2024] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
The European Union-sponsored ONCOBIOME network has spurred an international effort to identify and validate relevant gut microbiota-related biomarkers in oncology, generating a unique and publicly available microbiome resource. ONCOBIOME explores the effects of the microbiota on gut permeability and metabolism as well as on antimicrobial and antitumor immune responses. Methods for the diagnosis of gut dysbiosis have been developed based on oncomicrobiome signatures associated with the diagnosis, prognosis and treatment responses in patients with cancer. The mechanisms explaining how dysbiosis compromises natural or therapy-induced immunosurveillance have been explored. Through its integrative approach of leveraging multiple cohorts across populations, cancer types and stages, ONCOBIOME has laid the theoretical and practical foundations for the recognition of microbiota alterations as a hallmark of cancer. ONCOBIOME has launched microbiota-centered interventions and lobbies in favor of official guidelines for avoiding diet-induced or iatrogenic (for example, antibiotic- or proton pump inhibitor-induced) dysbiosis. Here, we review the key advances of the ONCOBIOME network and discuss the progress toward translating these into oncology clinical practice.
Collapse
Affiliation(s)
- Laurence Zitvogel
- INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
- Clinicobiome, Gustave Roussy, Villejuif, France.
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Clinicobiome, Gustave Roussy, Villejuif, France
| | - Bertrand Routy
- University of Montreal Research Center (CR-CHUM), Montreal, Quebec, Canada
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Sibylle Loibl
- German Breast Group c/ GBG Forschungs GmbH, Neu-Isenburg, Goethe University, Frankfurt, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - I Jolanda M de Vries
- Medical Biosciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars Engstrand
- Department of Microbiology Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology IRCCS, Milan, Italy
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
35
|
Chalif J, Goldstein N, Mehra Y, Spakowicz D, Chambers LM. The Role of the Microbiome in Cancer Therapies: Current Evidence and Future Directions. Hematol Oncol Clin North Am 2025; 39:269-294. [PMID: 39856008 DOI: 10.1016/j.hoc.2024.12.005] [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] [Indexed: 01/27/2025]
Abstract
The microbiome is essential for maintaining human health and is also a key factor in the development and progression of various diseases, including cancer. Growing evidence has highlighted the microbiome's significant impact on cancer development, progression, and treatment outcomes. As research continues to unfold, the microbiome and its modulation stand out as a promising frontier in cancer research and therapy. This review highlights current literature on the interplay between various cancer treatment modalities and human microbiotas, focusing on how the microbiome may affect treatment efficacy and toxicity and its potential as a therapeutic target to enhance future outcomes.
Collapse
Affiliation(s)
- Julia Chalif
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Naomi Goldstein
- Division of Obstetrics & Gynecology, The Ohio State University, Columbus, OH, USA
| | - Yogita Mehra
- Department of Medical Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Dan Spakowicz
- Department of Medical Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Laura M Chambers
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA.
| |
Collapse
|
36
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [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: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
37
|
Zugman M, Wong M, Jaime-Casas S, Pal SK. The gut microbiome and dietary metabolites in the treatment of renal cell carcinoma. Urol Oncol 2025; 43:244-253. [PMID: 39095306 DOI: 10.1016/j.urolonc.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
The gut microbiome is interlinked with renal cell carcinoma (RCC) and its response to systemic treatment. Mounting data suggests that certain elements of the gut microbiome may correlate with improved outcomes. New generation sequencing techniques and advanced bioinformatic data curation are accelerating the investigation of specific markers and metabolites that could predict treatment response. A variety of new therapeutic strategies, such as fecal microbiota transplantation, probiotic supplements, and dietary interventions, are currently being developed to modify the gut microbiome and improve anticancer therapies in patients with RCC. This review discusses the preliminary evidence indicating the role of the microbiome in cancer treatment, the techniques and tools necessary for its proper study and some of the current forms with which the microbiome can be modulated to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Zugman
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA; Centro de Oncologia e Hematologia Família Dayan-Daycoval Einstein, Hospital Israelita Albert, São Paulo, São Paulo, Brazil
| | - Megan Wong
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Salvador Jaime-Casas
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA.
| |
Collapse
|
38
|
Alves Costa Silva C, Almonte AA, Zitvogel L. Oncobiomics: Leveraging Microbiome Translational Research in Immuno-Oncology for Clinical-Practice Changes. Biomolecules 2025; 15:504. [PMID: 40305219 PMCID: PMC12024955 DOI: 10.3390/biom15040504] [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: 01/26/2025] [Revised: 03/16/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Growing evidence suggests that cancer should not be viewed solely as a genetic disease but also as the result of functional defects in the metaorganism, including disturbances in the gut microbiota (i.e., gut dysbiosis). The human microbiota plays a critical role in regulating epithelial barrier function in the gut, airways, and skin, along with host metabolism and systemic immune responses against microbes and cancer. Collaborative international networks, such as ONCOBIOME, are essential in advancing research equity and building microbiome resources to identify and validate microbiota-related biomarkers and therapies. In this review, we explore the intricate relationship between the microbiome, metabolism, and cancer immunity, and we propose microbiota-based strategies to improve outcomes for individuals at risk of developing cancer or living with the disease.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
| | - Andrew A. Almonte
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, 94805 Villejuif, France
| |
Collapse
|
39
|
Peng K, Li Y, Yang Q, Yu P, Zeng T, Lin C, Deng Y, Chen J. The therapeutic promise of probiotic Bacteroides fragilis (BF839) in cancer immunotherapy. Front Microbiol 2025; 16:1523754. [PMID: 40231233 PMCID: PMC11995047 DOI: 10.3389/fmicb.2025.1523754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Background Overwhelming evidence suggests that the gut microbiota modulates tumor response to immune checkpoint inhibitors (ICIs). The probiotic Bacteroides fragilis (BF839) was extensively used in China to improve gut microbiota dysbiosis-related symptoms. We hypothesized that probiotic BF839 could enhance tumor sensitivity to ICIs. Methods In the preclinical studies, mice received BF839 orally, PD-1 intraperitoneal injection, or a combination therapy of the two agents. The antitumor effect of BF839 was investigated by assessing the tumor growth and tumor immune microenvironment. Mice fecal samples were collected for 16S rRNA sequencing. Fresh tumor samples were collected for 16S RNA sequencing. The data of 29 patients with advanced solid tumor who received BF839 adjuvant therapy were retrospectively evaluated. The primary endpoint was overall survival (OS). Results Among patients with advanced solid tumors undergoing ICIs and chemotherapy, patients in BF839 long-term adjuvant treatment group had longer OS (p = 0.0101) than the BF839 short-term adjuvant treatment group. In the preclinical studies, we found that monotherapy with BF839 or anti-PD-1 antibody significantly inhibit tumor growth. Interestingly, BF839 worked synergistically with anti-PD-1 antibody and induced tumor regression, mediated by increased CD8+T cell infiltration. Mechanistically, BF839 induced tumor suppression was regulated by the cGAS-STING pathway. 16S rRNA sequencing results of mice fecal samples showed that BF839 treatment increased gut microbiota diversity. Conclusion Overall, our data suggest that BF839 enhanced tumor sensitivity to ICIs through cGAS-STING signaling. In the future, the application of probiotic BF839 to regulate gut microbiota may be a new strategy to enhance the efficacy of ICIs.
Collapse
Affiliation(s)
- Kunwei Peng
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuqing Li
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qijun Yang
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Peijin Yu
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ting Zeng
- Department of Clinical Nutrition, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chuhui Lin
- Department of Clinical Nutrition, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuhong Deng
- Department of Clinical Nutrition, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingqi Chen
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
40
|
Stanton SE, Anderson KG, Bruno TC, Capitini CM, Disis ML, McQuade J, Radvanyi L, Vanpouille-Box C, Wargo J, Baines KJ, Hong MMY, Rajeh A, Kim RH, Awadalla P, Hughes LK, Maleki Vareki S. SITC strategic vision: prevention, premalignant immunity, host and environmental factors. J Immunother Cancer 2025; 13:e010419. [PMID: 40154956 PMCID: PMC11956356 DOI: 10.1136/jitc-2024-010419] [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: 08/21/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Cancer immunotherapy has improved the survival of a subset of patients by harnessing the power of the immune system to find and destroy malignant cells. The immune system also protects the host by destroying developing premalignant and malignant tumors. Advancing our knowledge of premalignant immunity and immune changes seen in lesions that develop into invasive cancer versus those that regress offers an exciting opportunity to leverage the immune system for immune prevention and immune interception of premalignancy. Understanding the immune environment of premalignant lesions and how chronic inflammation plays a central role in the evolution of premalignancy is essential for developing effective immunoprevention and immune interceptions. Factors such as host genomics and environmental factors that affect premalignant immunity and the outcome of advanced cancers are equally important in determining the response to immunotherapy. The broad use of antibiotics and factors such as obesity can disrupt a healthy gut microbiome and drive chronic inflammation that suppresses preventive immunity or the antitumor immune response required for successful immunotherapy in advanced cancers. Modifiable lifestyle factors such as diet, obesity, smoking, and stress should be considered in designing immune prevention and interception studies, as well as for patients who receive immunotherapy for advanced cancer treatment. Other factors, such as the overall immune health of patients and existing comorbidities, affect both premalignant immunity and response to immunotherapy and, therefore, should be considered in managing patients with or without cancer. The Society for Immunotherapy of Cancer previously developed an overarching manuscript regarding the challenges and opportunities that exist in cancer immunotherapy, and this manuscript serves as an in-depth follow-up regarding the topics of premalignant immunity, immune interception, and immunoprevention, and the impact of the host on responding to immunotherapy.
Collapse
Affiliation(s)
- Sasha E Stanton
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Kristin G Anderson
- Department of Microbiology, Immunology and Cancer Biology, Department of Obstetrics and Gynecology, Beirne B. Carter Center for Immunology Research and the University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, Virginia, UK
| | - Tullia C Bruno
- Department of Immunology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christian M Capitini
- Department of Pediatrics and Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Mary L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington, USA
| | - Jennifer McQuade
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laszlo Radvanyi
- Ontario Institute for Cancer Research and Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, New York, New York, USA
| | - Jennifer Wargo
- Departments of Surgical Oncology and Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kelly J Baines
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Megan M Y Hong
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Adnan Rajeh
- Department of Oncology, Western University, London, Ontario, Canada
| | - Raymond H Kim
- Ontario Institute for Cancer Research and Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Familial Cancer Clinic, Princess Margaret Hospital Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Phillip Awadalla
- Ontario Institute for Cancer Research and Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Lauren K Hughes
- Ontario Institute for Cancer Research and Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Saman Maleki Vareki
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
41
|
Zhang S, Chen W, Zhou J, Liang Q, Zhang Y, Su M, Zhang Z, Qu J. The Benefits and Safety of Monoclonal Antibodies: Implications for Cancer Immunotherapy. J Inflamm Res 2025; 18:4335-4357. [PMID: 40162076 PMCID: PMC11952073 DOI: 10.2147/jir.s499403] [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: 10/05/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Monoclonal antibodies (mAbs) have transformed cancer treatment by providing highly targeted and effective therapies that specifically attack cancer cells, thus reducing the likelihood of adverse events (AEs) in patients. mAbs exert their action through various mechanisms, such as receptor blockade, antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and inhibition of immune checkpoints (eg, PD-1, PD-L1, and CTLA-4). These therapies have led to significant improvements in the treatment of several cancers, including HER2-positive breast cancer, non-small cell lung cancer (NSCLC), and melanoma. The efficacy of mAb therapy in cancer treatment is influenced by various intrinsic and extrinsic factors, such as environmental exposures, psychosocial factors, infection status, ways of life, and tumor microenvironment (TME), all of which can impact immune responses and treatment outcomes. Notably, the therapeutic benefits of mAbs are often accompanied by immune-related AEs (irAEs), which can vary from mild to severe and affect multiple organ systems. The dual nature of mAbs-stimulating antitumor immune responses while also inducing immune-related side effects-presents a notable challenge in clinical practice. This review highlights the importance of proactive strategies for managing irAEs, such as early detection, corticosteroid use, targeted immunosuppressive treatments, and the urgent need for reliable predictive biomarkers to improve treatment outcomes. Advancements in the prevention, prediction, and management of irAEs are essential to enhance the safety and effectiveness of mAb-based therapies, ultimately aiming to improve cancer patient outcomes.
Collapse
Affiliation(s)
- Shuguang Zhang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Wenying Chen
- Department of Pharmacy, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Jihong Zhou
- Department of Respiratory and Critical Care Medicine, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Qi Liang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Yu Zhang
- Department of Intensive Care Unit, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Ming Su
- Department of Respiratory and Critical Care Medicine, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Zilong Zhang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Jian Qu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, People’s Republic of China
| |
Collapse
|
42
|
Wong MK, Boukhaled GM, Armstrong E, Liu R, Heirali AA, Yee NR, Tsang J, Spiliopoulou P, Schneeberger PHH, Wang BX, Cochrane K, Sherriff K, Allen-Vercoe E, Siu LL, Spreafico A, Coburn B. Microbial Ecosystem Therapeutics 4 (MET4) elicits treatment-specific IgG responses associated with changes in gut microbiota in immune checkpoint inhibitor recipients with advanced solid tumors. J Immunother Cancer 2025; 13:e010681. [PMID: 40121033 PMCID: PMC11979602 DOI: 10.1136/jitc-2024-010681] [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: 09/26/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Gut microbiome modulation has shown promise in its potential to treat cancer in combination with immunotherapy. Mechanistically, the pathways and routes by which gut microbiota may influence systemic and antitumor immunity remain uncertain. Here, we used blood and stool samples from Microbial Ecosystem Therapeutic 4 (MET4)-IO, an early-phase trial testing the safety and engraftment of the MET4 bacterial consortium in immune checkpoint inhibitor recipients, to assess how MET4 may affect systemic immunity. METHODS Circulating antibody responses induced by MET4 were assessed using an antimicrobial antibody flow cytometry assay on pretreatment and post-treatment plasma. Antibody responses were associated with taxonomic changes in stool identified by metagenomic sequencing. Mass cytometry was performed on peripheral blood mononuclear cells to identify shifts in circulating immune subsets associated with antibody responses. RESULTS Increases in circulating anti-MET4 immunoglobulin G (IgG) responses were measured by flow cytometry post-consortium treatment in MET4 recipients, but not untreated control participants, with five individuals displaying notably higher antibody responses. Stronger IgG responses were associated with greater increases in multiple taxa, including MET4 microbe Collinsella aerofaciens, which was previously linked with immune checkpoint response. However, these taxa were not enriched in the IgG-bound fraction post-MET4 treatment. Greater increases in circulating B cells and FoxP3+ CD4+ T cells post-MET4 treatment were observed in the blood of high IgG responders, while CD14+ and CD16+ monocyte populations were decreased in these individuals. CONCLUSION These results demonstrate the induction of treatment-specific circulating humoral immunity by a bacterial consortium and suggest potential mechanisms by which gut microbes may contribute to antitumor immunity.
Collapse
Affiliation(s)
- Matthew K Wong
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Giselle M Boukhaled
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eric Armstrong
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Rachel Liu
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Alya A Heirali
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Noelle R Yee
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jinny Tsang
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Pavlina Spiliopoulou
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Pierre H H Schneeberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Ben X Wang
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | | | - Lillian L Siu
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Anna Spreafico
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Bryan Coburn
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Infectious Diseases, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Wilson RP, Rink L, Tükel Ç. Microbiota and cancer: unraveling the significant influence of microbial communities on cancer treatment. Cancer Metastasis Rev 2025; 44:42. [PMID: 40120010 DOI: 10.1007/s10555-025-10256-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Affiliation(s)
- R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Lori Rink
- Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
- Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Zhao S, Lu Z, Zhao F, Tang S, Zhang L, Feng C. Assessing the impact of probiotics on immunotherapy effectiveness and antibiotic-mediated resistance in cancer: a systematic review and meta-analysis. Front Immunol 2025; 16:1538969. [PMID: 40191197 PMCID: PMC11968366 DOI: 10.3389/fimmu.2025.1538969] [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: 12/03/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
Background Probiotics have been demonstrated to exert a potential clinical enhancing effect in cancer patients receiving immune checkpoint inhibitors (ICIs), while antibiotics exert a detrimental impact. Prior meta-analysis papers have substantial limitations and are devoid of recent published studies. Therefore, this study aimed to perform an updated meta-analysis and, for the first time, assess whether probiotics can restore the damage of antibiotics to immunotherapy. Methods A comprehensive literature search was conducted in three English databases and three Chinese databases with a cutoff date of August 11, 2024. The methodological quality of the studies was evaluated using the Newcastle-Ottawa Quality Assessment Scale (NOS) or the Revised Cochrane risk-of-bias tool (RoB 2). Engauge Digitizer v12.1 was employed to extract hazard ratios (HRs) with 95% confidence interval (CI) for survival outcomes when these data were not explicitly provided in the manuscripts. Meta-analysis was conducted using Stata 14 software. Results The study sample comprised eight retrospective and four prospective studies, involving a total of 3,142 participants. The findings indicate that probiotics significantly prolong the overall survival (OS) (I2 = 31.2%; HR=0.58, 95% CI: 0.46-0.73, p < 0.001) and progression-free survival (PFS) (I2 = 65.2%; HR=0.66, 95% CI: 0.54-0.81, p < 0.001) in cancer patients receiving ICIs, enhance the objective response rate (ORR) (I2 = 33.5%; OR=1.75, 95% CI: 1.27-2.40, p = 0.001) and disease control rate (DCR) (I2 = 50.0%; OR=1.93, 95% CI: 1.11-3.35, p = 0.002). For non-small cell lung cancer (NSCLC) patients exposed to antibiotics, the use of probiotics was associated with superior OS (I2 = 0.0%; HR=0.45, 95% CI: 0.34-0.59, p < 0.001) and PFS (I2 = 0.0%; HR=0.48, 95% CI: 0.38-0.62, p < 0.001) when compared to non-users. Subgroup differences were observed regarding the cancer type (P=0.006) and ethnic backgrounds (P=0.011) in OS. Conclusions The meta-analysis findings suggest that probiotics can effectively extend the survival of cancer treated with ICIs. In NSCLC, probiotics appear to mitigate the negative impact of antibiotics on immunotherapy effectiveness, which has profound clinical significance. Nevertheless, additional large-scale, high-quality randomized controlled trials are necessary to further validate these findings. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=579047, identifier CRD42024579047.
Collapse
Affiliation(s)
- Shuya Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Peking University People’s Hospital, Beijing, China
| | - Zian Lu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fangmin Zhao
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shihuan Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing, China
| | - Lishan Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Cuiling Feng
- Peking University People’s Hospital, Beijing, China
| |
Collapse
|
45
|
Chatrizeh M, Tian J, Rogers M, Feturi F, Wu G, Firek B, Nikonov R, Cass L, Sheppeck A, Ramos-Jiménez RG, Ohja L, Caroll A, Henkel M, Azar J, Aneja RK, Campfield B, Simon D, Morowitz MJ. Plant based enteral nutrition outperforms artificial nutrition in mitigating consequences of antibiotic-induced dysbiosis in mice and humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.19.25323813. [PMID: 40166543 PMCID: PMC11957089 DOI: 10.1101/2025.03.19.25323813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Malnutrition, gut inflammation, and antibiotic induced dysbiosis (AID) are omnipresent risk factors for poor clinical outcomes among critically ill patients. We previously showed that commercially available plant-based enteral nutrition (PBEN) preserves a commensal microbiome when compared to commonly used forms of commercially available artificial enteral nutrition (AEN). This study reveals that PBEN is superior to artificial enteral nutrition (AEN) in recovering from antibiotic-induced dysbiosis (AID) in mice and humans. PBEN effectively mitigates anemia, leukopenia, restores naïve lymphocyte populations, and reduces bone marrow myeloid cell expansion. Animals randomized to PBEN also fared better in response to infectious challenges after antibiotics. A pilot clinical study validated these findings, showing increased gut commensals, reduced pathogens, and improved leukocyte balance in critically ill patients receiving PBEN compared to AEN. These results suggest PBEN offers a practical dietary approach to mitigate antibiotic-associated complications and improve clinical outcomes among hospitalized patients requiring supplemental nutrition.
Collapse
Affiliation(s)
- Mona Chatrizeh
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jianmin Tian
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew Rogers
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Firuz Feturi
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guojun Wu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brian Firek
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roman Nikonov
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Cass
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandra Sheppeck
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Lavnish Ohja
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ali Caroll
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mathew Henkel
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Azar
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rajesh K Aneja
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian Campfield
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dennis Simon
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pediatric Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael J Morowitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Lead contact
| |
Collapse
|
46
|
Han Y, Guo X, Ji Z, Guo Y, Ma W, Du H, Guo Y, Xiao H. Colon health benefits of plant-derived exosome-like nanoparticles via modulating gut microbiota and immunity. Crit Rev Food Sci Nutr 2025:1-21. [PMID: 40105379 DOI: 10.1080/10408398.2025.2479066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Exosomes are nanoscale particles with a lipid bilayer membrane that were first identified in mammalian cells. Plant-derived exosome-like nanoparticles (PELNs) share structural and molecular similarities with mammalian exosomes, including lipids, proteins, microRNA (miRNA), and plant-derived metabolites. Owing to their unique characteristics, such as outstanding stability, low immunogenicity, high biocompatibility, and sustainability, PELNs have emerged as promising natural bioactive agents with the capacity for cross-kingdom cellular regulation. Dietary supplementation with PELNs, particularly from fruits and vegetables, has demonstrated health benefits. An increasing number of studies have indicated the beneficial effects of PELNs on colon health. This review summarizes the isolation and characterization of PELNs, and their stability, uptake, and distribution after oral ingestion. Furthermore, this review emphasizes the interactions between PELNs, gut microbiota, and the gut immune system, including the uptake of PELNs by gut microbiota, modulation of gut bacteria metabolism, and immune responses by PELNs. Additionally, the applications of PELNs as bioactive components and drug carriers targeting the colon are reviewed. In summary, PELNs represent a versatile and natural approach to improve colon health, with potential applications in both therapeutic and preventive healthcare strategies.
Collapse
Affiliation(s)
- Yanhui Han
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Xiaojing Guo
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Zhengmei Ji
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
| | - Yuxin Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Wenjun Ma
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Yurong Guo
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
47
|
Xiao Y, Yang Y, Gao S, Zhang H, Wang J, Lin T, Bai Y. Dietary index for gut microbiota, a novel protective factor for the prevalence of chronic kidney diseases in the adults: insight from NHANES 2007-2018. Front Nutr 2025; 12:1561235. [PMID: 40177175 PMCID: PMC11963806 DOI: 10.3389/fnut.2025.1561235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction This study explore the association between the dietary index for gut microbiota (DI-GM) and the prevalence of chronic kidney disease (CKD). Method A cross-sectional study of participants aged ≥20 years using the data drawn from NHANES (2007-2018). DI-GM is comprised 14 dietary components (10 beneficial and 4 unfavorable). CKD diagnosis based on urine albumin-to-creatinine ratio (uACR) and estimated glomerular filtration rate (eGFR). Logistic regression models were employed to evaluate the relationship between DI-GM and CKD while controlling for various covariates. Additionally, a spline smooth analysis was performed. Subgroup and interaction analyses were conducted to investigate whether any factors modified this relationship. Results A total of 28,843 participants were eligible for the study, of whom 5,461 were diagnosed with CKD, while 23,382 were not. Patients with CKD exhibited significantly lower DI-GM scores compared to healthy individuals. A negative association between DI-GM and the prevalence of CKD was observed across all models, with the relationship being more pronounced in individuals with DI-GM scores greater than 5 compared to those with scores ≤3. Beneficial components, such as dietary fiber, whole grains, and coffee, were identified as protective factors. Moreover, sex make an effect on this relationship, with stronger effects noted in women. Conclusion Higher DI-GM scores correlate with reduced CKD prevalence, and the effect appears to be more pronounced in women than in men. These findings suggest that enhancing gut health through diet may serve as a viable strategy for the prevention and management of CKD, with particular attention to sex-based differences in prevention.
Collapse
Affiliation(s)
- Yunfei Xiao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaqing Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shunyu Gao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jia Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Lin
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunjin Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Imyanitov EN, Preobrazhenskaya EV, Mitiushkina NV. Overview on biomarkers for immune oncology drugs. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002298. [PMID: 40135049 PMCID: PMC11933888 DOI: 10.37349/etat.2025.1002298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) are widely used in clinical oncology, less than half of treated cancer patients derive benefit from this therapy. Both tumor- and host-related variables are implicated in response to ICIs. The predictive value of PD-L1 expression is confined only to several cancer types, so this molecule is not an agnostic biomarker. Highly elevated tumor mutation burden (TMB) caused either by excessive carcinogenic exposure or by a deficiency in DNA repair is a reliable indicator for ICI efficacy, as exemplified by tumors with high-level microsatellite instability (MSI-H). Other potentially relevant tumor-related characteristics include gene expression signatures, pattern of tumor infiltration by immune cells, and, perhaps, some immune-response modifying somatic mutations. Host-related factors have not yet been comprehensively considered in relevant clinical trials. Microbiome composition, markers of systemic inflammation [e.g., neutrophil-to-lymphocyte ratio (NLR)], and human leucocyte antigen (HLA) diversity may influence the efficacy of ICIs. Studies on ICI biomarkers are likely to reveal modifiable tumor or host characteristics, which can be utilized to direct the antitumor immune defense. Examples of the latter approach include tumor priming to immune therapy by cytotoxic drugs and elevation of ICI efficacy by microbiome modification.
Collapse
Affiliation(s)
- Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
| |
Collapse
|
49
|
Lin Y, Xie M, Lau HCH, Zeng R, Zhang R, Wang L, Li Q, Wang Y, Chen D, Jiang L, Damsky W, Yu J. Effects of gut microbiota on immune checkpoint inhibitors in multi-cancer and as microbial biomarkers for predicting therapeutic response. MED 2025; 6:100530. [PMID: 39515321 DOI: 10.1016/j.medj.2024.10.007] [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: 10/04/2023] [Revised: 04/16/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Gut bacteria are related to immune checkpoint inhibitors (ICIs). However, there is inconsistency in ICI-associated species, while the role of non-bacterial microbes in immunotherapy remains elusive. Here, we evaluated the association of trans-kingdom microbes with ICIs by multi-cohort multi-cancer analyses. METHODS We retrieved fecal metagenomes from 1,359 ICI recipients with four different cancers (metastatic melanoma [MM], non-small cell lung carcinoma [NSCLC], renal cell cancer [RCC], and hepatocellular carcinoma) from 12 published datasets. Microbiota composition was analyzed using the Wilcoxon rank test. The performance of microbial biomarkers in predicting ICI response was assessed by random forest. Key responder-associated microbes were functionally examined in vitro and in mice. FINDINGS Trans-kingdom gut microbiota (bacteria, eukaryotes, viruses, and archaea) was significantly different between ICI responders and non-responders in multi-cancer. Bacteria (Faecalibacterium prausnitzii, Coprococcus comes) and eukaryotes (Nemania serpens, Hyphopichia pseudoburtonii) were consistently enriched in responders of ≥2 cancer types or from ≥3 cohorts, contrasting with the depleted bacterium Hungatella hathewayi. Responder-associated species in each cancer were revealed, such as F. prausnitzii in MM and 6 species in NSCLC. These signature species influenced ICI efficacy by modulating CD8+ T cell activity in vitro and in mice. Moreover, bacterial and eukaryotic biomarkers showed great performance in predicting ICI response in patients from discovery and two validation cohorts (MM: area under the receiver operating characteristic curve [AUROC] = 72.27%-80.19%; NSCLC: AUROC = 72.70%-87.98%; RCC: AUROC = 83.33%-89.58%). CONCLUSIONS This study identified trans-kingdom microbial signatures associated with ICI in multi-cancer and specific cancer types. Trans-kingdom microbial biomarkers are potential predictors of ICI response in patients with cancer. FUNDING Funding information is shown in the acknowledgments.
Collapse
Affiliation(s)
- Yufeng Lin
- Institute of Digestive Disease, 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 SAR, China
| | - Mingxu Xie
- Institute of Digestive Disease, 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 SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, 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 SAR, China
| | - Ruijie Zeng
- Institute of Digestive Disease, 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 SAR, China
| | - Ruyi Zhang
- Institute of Digestive Disease, 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 SAR, China
| | - Luyao Wang
- Institute of Digestive Disease, 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 SAR, China
| | - Qing Li
- Institute of Digestive Disease, 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 SAR, China; Department of Anaesthesia and Intensive Care, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yiwei Wang
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Danyu Chen
- Institute of Digestive Disease, 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 SAR, China
| | - Lanping Jiang
- Institute of Digestive Disease, 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 SAR, China
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jun Yu
- Institute of Digestive Disease, 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 SAR, China.
| |
Collapse
|
50
|
Sun J, Song S, Liu J, Chen F, Li X, Wu G. Gut microbiota as a new target for anticancer therapy: from mechanism to means of regulation. NPJ Biofilms Microbiomes 2025; 11:43. [PMID: 40069181 PMCID: PMC11897378 DOI: 10.1038/s41522-025-00678-x] [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: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
In order to decipher the relationship between gut microbiota imbalance and cancer, this paper reviewed the role of intestinal microbiota in anticancer therapy and related mechanisms, discussed the current research status of gut microbiota as a biomarker of cancer, and finally summarized the reasonable means of regulating gut microbiota to assist cancer therapy. Overall, our study reveals that the gut microbiota can serve as a potential target for improving cancer management.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shiyan Song
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiahua Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Xiaorui Li
- Department of oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|