1
|
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
|
2
|
Pezeshki B, Abdulabbas HT, Alturki AD, Mansouri P, Zarenezhad E, Nasiri-Ghiri M, Ghasemian A. Synergistic Interactions Between Probiotics and Anticancer Drugs: Mechanisms, Benefits, and Challenges. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10462-0. [PMID: 39873952 DOI: 10.1007/s12602-025-10462-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Research into the role of probiotics-often referred to as "living supplements"-in cancer therapy is still in its early stages, and uncertainties regarding their effectiveness remain. Relevantly, chemopreventive and therapeutic effects of probiotics have been determined. There is also substantial evidence supporting their potential in cancer treatment such as immunotherapy. Probiotics employ various mechanisms to inhibit cancer initiation and progression. These include colonizing and protecting the gastrointestinal tract (GIT), producing metabolites, inducing apoptosis and autophagy, exerting anti-inflammatory properties, preventing metastasis, enhancing the effectiveness of immune checkpoint inhibitors (ICIs), promoting cancer-specific T cell infiltration, arresting the cell cycle, and exhibiting direct or indirect synergistic effects with anticancer drugs. Additionally, probiotics have been shown to activate tumor suppressor genes and inhibit pro-inflammatory transcription factors. They also increase reactive oxygen species production within cancer cells. Synergistic interactions between probiotics and various anticancer drugs, such as cisplatin, cyclophosphamide, 5-fluorouracil, trastuzumab, nivolumab, ipilimumab, apatinib, gemcitabine, tamoxifen, sorafenib, celecoxib and irinotecan have been observed. The combination of probiotics with anticancer drugs holds promise in overcoming drug resistance, reducing recurrence, minimizing side effects, and lowering treatment costs. In addition, fecal microbiota transplantation (FMT) and prebiotics supplementation has increased cytotoxic T cells within tumors. However, probiotics may leave some adverse effects such as risk of infection and gastrointestinal effects, antagonistic effects with drugs, and different responses among patients. These findings highlight insights for considering specific strains and engineered probiotic applications, preferred doses and timing of treatment, and personalized therapies to enhance the efficacy of cancer therapy. Accordingly, targeted interventions and guidelines establishment needs extensive randomized controlled trials as probiotic-based cancer therapy has not been approved by Food and Drug Administration (FDA).
Collapse
Affiliation(s)
- Babak Pezeshki
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Hussein T Abdulabbas
- Department of Medical Microbiology, Medical College, Al Muthanna University, Samawah, Al Muthanna, Iraq
| | - Ahmed D Alturki
- Department of Medical Laboratories Techniques, Imam Ja'afar Al-Sadiq University, Samawah, Al-Muthanna, Iraq
| | - Pegah Mansouri
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahdi Nasiri-Ghiri
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
3
|
Le Ngoc K, Pham TTH, Nguyen TK, Huong PT. Pharmacomicrobiomics in precision cancer therapy: bench to bedside. Front Immunol 2024; 15:1428420. [PMID: 39315107 PMCID: PMC11416994 DOI: 10.3389/fimmu.2024.1428420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The burgeoning field of pharmacomicrobiomics offers promising insights into the intricate interplay between the microbiome and cancer, shaping responses to diverse treatment modalities. This review aims to analyze the molecular mechanisms underlying interactions between distinct microbiota types and cancer, as well as their influence on treatment outcomes. We explore how the microbiome impacts antitumor immunity, and response to chemotherapy, immunotherapy, and radiation therapy, unveiling its multifaceted roles in cancer progression and therapy resistance. Moreover, we discuss the challenges hindering the development of microbiome-based interventions in cancer therapy, including standardization, validation, and clinical translation. By synthesizing clinical evidence, we underscore the transformative potential of harnessing pharmacomicrobiomics in guiding cancer treatment decisions, paving the way for improved patient outcomes in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Phung Thanh Huong
- Faculty of Biotechnology, Hanoi University of Pharmacy,
Hanoi, Vietnam
| |
Collapse
|
4
|
Dong Y, Wei J, Yang F, Qu Y, Huang J, Shi D. Nutrient-Based Approaches for Melanoma: Prevention and Therapeutic Insights. Nutrients 2023; 15:4483. [PMID: 37892558 PMCID: PMC10609833 DOI: 10.3390/nu15204483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Melanoma, a prevalent and lethal form of skin cancer, remains a formidable challenge in terms of prevention and treatment. While significant progress has been made in understanding its pathogenesis and treatment, the quest for effective prevention strategies and therapeutic approaches remains ongoing. Considering the increased advancements in understanding the dynamic interplay between nutrients and melanoma, we aim to offer a refreshed perspective on nutrient-based approaches for melanoma prevention and adjunctive therapy. In contrast to other studies, we have innovatively provided a detailed exposition of the nutrients' influences on melanoma prognosis and treatment. This review firstly examines various nutrients, including antioxidants (namely vitamins A, D, C, and E; selenium; and caffeine), polyunsaturated fatty acids, and flavonoids, for their effects and underlying mechanisms in reducing melanoma risk. Among these nutrients, caffeine shows the most promising potential, as it is supported by multiple cohort studies for its protective effect against melanoma. In contrast, there is a certain degree of inconsistency in the research of other nutrients, possibly due to inherent differences between animal studies and epidemiological research, as well as variations in the definition of nutrient intake. To comprehensively investigate the impact of nutrients on melanoma progression and therapeutic approaches, the following sections will explore how nutrients influence immune responses and other physiological processes. While there is robust support from cell and animal studies regarding the immunomodulatory attributes of vitamins D and zinc, the anti-angiogenic potential of polyphenols, and the cell growth-inhibitory effects of flavonoids, the limited availability of human-based research substantially constrains their practical relevance in clinical contexts. As for utilizing nutrients in adjuvant melanoma treatments, multiple approaches have garnered clinical research support, including the utilization of vitamin D to decrease the postoperative recurrence rates among melanoma patients and the adoption of a high-fiber diet to enhance the effectiveness of immunotherapy. In general, the effects of most nutrients on reducing the risk of melanoma are not entirely clear. However, several nutrients, including vitamin D and dietary fiber, have demonstrated their potential to improve the melanoma prognosis and enhance the treatment outcomes, making them particularly deserving of clinical attention. A personalized and interdisciplinary approach, involving dermatologists, oncologists, nutritionists, and researchers, holds the promise of optimizing melanoma treatment strategies.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Yang Qu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Di Shi
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| |
Collapse
|
5
|
Boesch M, Baty F, Rassouli F, Kowatsch T, Joerger M, Früh M, Brutsche MH. Non-pharmaceutical interventions to optimize cancer immunotherapy. Oncoimmunology 2023; 12:2255459. [PMID: 37791231 PMCID: PMC10543347 DOI: 10.1080/2162402x.2023.2255459] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023] Open
Abstract
The traditional picture of cancer patients as weak individuals requiring maximum rest and protection is beginning to dissolve. Too much focus on the medical side and one's own vulnerability and mortality might be counterproductive and not doing justice to the complexity of human nature. Unlike cytotoxic and lympho-depleting treatments, immune-engaging therapies strengthen the immune system and are typically less harmful for patients. Thus, cancer patients receiving checkpoint inhibitors are not viewed as being vulnerable per se, at least not in immunological and physical terms. This perspective article advocates a holistic approach to cancer immunotherapy, with an empowered patient in the center, focusing on personal resources and receiving domain-specific support from healthcare professionals. It summarizes recent evidence on non-pharmaceutical interventions to enhance the efficacy of immune checkpoint blockade and improve quality of life. These interventions target behavioral factors such as diet, physical activity, stress management, circadian timing of checkpoint inhibitor infusion, and waiving unnecessary co-medication curtailing immunotherapy efficacy. Non-pharmaceutical interventions are universally accessible, broadly applicable, instantly actionable, scalable, and economically sustainable, creating value for all stakeholders involved. Most importantly, this holistic framework re-emphasizes the patient as a whole and harnesses the full potential of anticancer immunity and checkpoint blockade, potentially leading to survival benefits. Digital therapeutics are proposed to accompany the patients on their mission toward change in lifestyle-related behaviors for creating optimal conditions for treatment efficacy and personal growth.
Collapse
Affiliation(s)
| | - Florent Baty
- Lung Center, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
| | - Frank Rassouli
- Lung Center, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
| | - Tobias Kowatsch
- Institute for Implementation Science in Health Care, University of Zurich, Zurich, Switzerland
- School of Medicine, University of St.Gallen, St.Gallen, Switzerland
- Centre for Digital Health Interventions, Department of Technology, Management, and Economics, ETH Zurich, Zurich, Switzerland
| | - Markus Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
| | - Martin Früh
- Department of Medical Oncology and Hematology, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
- Department of Medical Oncology, University Hospital Bern, Bern, Switzerland
| | | |
Collapse
|
6
|
Feng X, Li Z, Guo W, Hu Y. The effects of traditional Chinese medicine and dietary compounds on digestive cancer immunotherapy and gut microbiota modulation: A review. Front Immunol 2023; 14:1087755. [PMID: 36845103 PMCID: PMC9945322 DOI: 10.3389/fimmu.2023.1087755] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
Digestive tract-related cancers account for four of the top ten high-risk cancers worldwide. In recent years, cancer immunotherapy, which exploits the innate immune system to attack tumors, has led to a paradigm shifts in cancer treatment. Gut microbiota modification has been widely used to regulate cancer immunotherapy. Dietary compounds and traditional Chinese medicine (TCM) can alter the gut microbiota and its influence on toxic metabolite production, such as the effect of iprindole on lipopolysaccharide (LPS), and involvement in various metabolic pathways that are closely associated with immune reactions. Therefore, it is an effective strategy to explore new immunotherapies for gastrointestinal cancer to clarify the immunoregulatory effects of different dietary compounds/TCMs on intestinal microbiota. In this review, we have summarized recent progress regarding the effects of dietary compounds/TCMs on gut microbiota and their metabolites, as well as the relationship between digestive cancer immunotherapy and gut microbiota. We hope that this review will act as reference, providing a theoretical basis for the clinical immunotherapy of digestive cancer via gut microbiota modulation.
Collapse
Affiliation(s)
- Xiaoli Feng
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhenhao Li
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weihong Guo
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Weihong Guo, ; Yanfeng Hu,
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Weihong Guo, ; Yanfeng Hu,
| |
Collapse
|
7
|
Sheinboim D, Parikh S, Manich P, Markus I, Dahan S, Parikh R, Stubbs E, Cohen G, Zemser-Werner V, Bell RE, Ruiz SA, Percik R, Brenner R, Leibou S, Vaknine H, Arad G, Gerber Y, Keinan-Boker L, Shimony T, Bikovski L, Goldstein N, Constantini K, Labes S, Mordechai S, Doron H, Lonescu A, Ziv T, Nizri E, Choshen G, Eldar-Finkelman H, Tabach Y, Helman A, Ben-Eliyahu S, Erez N, Perlson E, Geiger T, Ben-Zvi D, Khaled M, Gepner Y, Levy C. An Exercise-Induced Metabolic Shield in Distant Organs Blocks Cancer Progression and Metastatic Dissemination. Cancer Res 2022; 82:4164-4178. [PMID: 36084256 PMCID: PMC9762351 DOI: 10.1158/0008-5472.can-22-0237] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/16/2022] [Accepted: 08/31/2022] [Indexed: 01/07/2023]
Abstract
Exercise prevents cancer incidence and recurrence, yet the underlying mechanism behind this relationship remains mostly unknown. Here we report that exercise induces the metabolic reprogramming of internal organs that increases nutrient demand and protects against metastatic colonization by limiting nutrient availability to the tumor, generating an exercise-induced metabolic shield. Proteomic and ex vivo metabolic capacity analyses of murine internal organs revealed that exercise induces catabolic processes, glucose uptake, mitochondrial activity, and GLUT expression. Proteomic analysis of routinely active human subject plasma demonstrated increased carbohydrate utilization following exercise. Epidemiologic data from a 20-year prospective study of a large human cohort of initially cancer-free participants revealed that exercise prior to cancer initiation had a modest impact on cancer incidence in low metastatic stages but significantly reduced the likelihood of highly metastatic cancer. In three models of melanoma in mice, exercise prior to cancer injection significantly protected against metastases in distant organs. The protective effects of exercise were dependent on mTOR activity, and inhibition of the mTOR pathway with rapamycin treatment ex vivo reversed the exercise-induced metabolic shield. Under limited glucose conditions, active stroma consumed significantly more glucose at the expense of the tumor. Collectively, these data suggest a clash between the metabolic plasticity of cancer and exercise-induced metabolic reprogramming of the stroma, raising an opportunity to block metastasis by challenging the metabolic needs of the tumor. SIGNIFICANCE Exercise protects against cancer progression and metastasis by inducing a high nutrient demand in internal organs, indicating that reducing nutrient availability to tumor cells represents a potential strategy to prevent metastasis. See related commentary by Zerhouni and Piskounova, p. 4124.
Collapse
Affiliation(s)
- Danna Sheinboim
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shivang Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Paulee Manich
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Markus
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Sapir Dahan
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roma Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elisa Stubbs
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Gali Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.,Stanley Steyer Institute for Cancer Epidemiology and Research, Tel Aviv University, Tel Aviv, Israel
| | | | - Rachel E. Bell
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Arciniegas Ruiz
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruth Percik
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Endocrinology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Ronen Brenner
- Institute of Oncology, E. Wolfson Medical Center, Holon, Israel
| | - Stav Leibou
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hananya Vaknine
- Institute of Pathology, E. Wolfson Medical Center, Holon, Israel
| | - Gali Arad
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yariv Gerber
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.,Stanley Steyer Institute for Cancer Epidemiology and Research, Tel Aviv University, Tel Aviv, Israel
| | - Lital Keinan-Boker
- School of Public Health, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel.,Israel Center for Disease Control, Israel Ministry of Health, Ramat Gan, Israel
| | - Tal Shimony
- Israel Center for Disease Control, Israel Ministry of Health, Ramat Gan, Israel
| | - Lior Bikovski
- The Myers Neuro-Behavioral Core Facility, Tel Aviv University, Tel Aviv, Israel.,School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| | - Nir Goldstein
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Keren Constantini
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Sapir Labes
- Department of Developmental Biology and Cancer Research, Institute of Medical Research-Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shimonov Mordechai
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Surgery, E. Wolfson Medical Center, Holon, Israel
| | - Hila Doron
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Lonescu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Ziv
- The Smoler Proteomics Center, Technion, Haifa, Israel
| | - Eran Nizri
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Dermatology, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
| | - Guy Choshen
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Internal Medicine, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
| | - Hagit Eldar-Finkelman
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Tabach
- Department of Developmental Biology and Cancer Research, Institute of Medical Research-Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aharon Helman
- Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, Israel
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Geiger
- The Weizmann Institute of Science, Rehovot, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, Institute of Medical Research Israel–Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mehdi Khaled
- INSERM 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Corresponding Authors: Carmit Levy, Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, 69978, Israel. E-mail: ; Yftach Gepner, E-mail: ; and Mehdi Khaled, E-mail:
| | - Yftach Gepner
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.,Corresponding Authors: Carmit Levy, Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, 69978, Israel. E-mail: ; Yftach Gepner, E-mail: ; and Mehdi Khaled, E-mail:
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Corresponding Authors: Carmit Levy, Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, 69978, Israel. E-mail: ; Yftach Gepner, E-mail: ; and Mehdi Khaled, E-mail:
| |
Collapse
|
8
|
Montégut L, de Cabo R, Zitvogel L, Kroemer G. Science-Driven Nutritional Interventions for the Prevention and Treatment of Cancer. Cancer Discov 2022; 12:2258-2279. [PMID: 35997502 PMCID: PMC10749912 DOI: 10.1158/2159-8290.cd-22-0504] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/18/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
In population studies, dietary patterns clearly influence the development, progression, and therapeutic response of cancers. Nonetheless, interventional dietary trials have had relatively little impact on the prevention and treatment of malignant disease. Standardization of nutritional interventions combined with high-level mode-of-action studies holds the promise of identifying specific entities and pathways endowed with antineoplastic properties. Here, we critically review the effects of caloric restriction and more specific interventions on macro- and micronutrients in preclinical models as well as in clinical studies. We place special emphasis on the prospect of using defined nutrition-relevant molecules to enhance the efficacy of established anticancer treatments. SIGNIFICANCE The avoidance of intrinsically hypercaloric and toxic diets contributes to the prevention and cure of cancer. In addition, specific diet-induced molecules such as ketone bodies and micronutrients, including specific vitamins, have drug-like effects that are clearly demonstrable in preclinical models, mostly in the context of immunotherapies. Multiple trials are underway to determine the clinical utility of such molecules.
Collapse
Affiliation(s)
- Léa Montégut
- Equipe labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland
| | - Laurence Zitvogel
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Gustave Roussy Comprehensive Cancer Institute, ClinicObiome, Villejuif, France
- INSERM U1015, Paris, France
- Equipe labellisée par la Ligue contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) BIOTHERIS, Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, 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
|
9
|
Dai Z, Fu J, Peng X, Tang D, Song J. Intestinal Microbiota: The Driving Force behind Advances in Cancer Immunotherapy. Cancers (Basel) 2022; 14:4796. [PMID: 36230724 PMCID: PMC9564057 DOI: 10.3390/cancers14194796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, cancer immunotherapy has become a breakthrough method to solve solid tumors. It uses immune checkpoint inhibitors to interfere with tumor immune escape to coordinate anti-tumor therapy. However, immunotherapy has an individualized response rate. Moreover, immune-related adverse events and drug resistance are still urgent issues that need to be resolved, which may be attributed to the immune imbalance caused by immune checkpoint inhibitors. Microbiome research has fully revealed the metabolic-immune interaction relationship between the microbiome and the host. Surprisingly, sequencing technology further proved that intestinal microbiota could effectively intervene in tumor immunotherapy and reduce the incidence of adverse events. Therefore, cancer immunotherapy under the intervention of intestinal microbiota has innovatively broadened the anti-tumor landscape and is expected to become an active strategy to enhance individualized responses.
Collapse
Affiliation(s)
- Zhujiang Dai
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai 200092, China
| | - Jihong Fu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai 200092, China
| | - Xiang Peng
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai 200092, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Jinglue Song
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai 200092, China
| |
Collapse
|