1
|
Zhao R, Cao B, Li H, Gao J, Xu Q, Cui H, Yuan Z, Ren H, Wei B. MZT1 protects gastric cancer against glucose starvation through targeting NEDD1. Life Sci 2025; 372:123622. [PMID: 40204068 DOI: 10.1016/j.lfs.2025.123622] [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: 09/12/2024] [Revised: 03/19/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
A fasting mimic diet (FMD) has been proven to be a potential therapeutic regimen for gastric cancer (GC) patients. However, the intolerance of energy restriction and limited efficacies hinder wide application of FMD. To identify critical targets mediating resistance against glucose starvation and explore novel approaches to GC therapy, proteomics profiling was performed to depict the landscape of protein expression changes in cells under glucose deprivation. MZT1 was found to be greatly upregulated. We next investigated potential clinical value and regulatory functions of MZT1. Compared to adjacent normal tissues, MZT1 was upregulated in GC specimens and associated with unfavorable patient prognosis. Both in vitro and in vivo experiments indicated that downregulation of the MZT1 level inhibited GC proliferation, migration, invasion, glycolysis and sensitized cells to glucose starvation. Mechanistically, MZT1 functioned as an oncogenic factor by inhibiting NEDD1 ubiquitination and increasing its expression. In conclusion, during glucose starvation, MZT1 is upregulated in GC cells, which promotes resistance by directly suppression of NEDD1 ubiquitination. Our findings unveil the novel mechanisms by which MZT1 can promote GC malignancy. The potential clinical value of MZT1 as GC biomarkers has been first revealed. Suppression of MZT1 may become a promising approach to improve FMD efficacy, which require further validation by future investigations.
Collapse
Affiliation(s)
- Ruiyang Zhao
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Bo Cao
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Hanghang Li
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Jingwang Gao
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Qixuan Xu
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Hao Cui
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Zhen Yuan
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Huiguang Ren
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Bo Wei
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
2
|
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] [MESH Headings] [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
|
3
|
Ye J, Hu P, Zhang R, Zhou L, Luo Z, Chen Y, Ruan S, Zhu M, Ding H, Qian Y, Xing Y, Meng T, Wang C, Song D. Targeting Hyperglycemic Bone Pre-Metastatic Niche for Breast Cancer Bone Metastasis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04924. [PMID: 40491413 DOI: 10.1002/advs.202504924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/26/2025] [Indexed: 06/11/2025]
Abstract
Bone is the most common site of breast cancer metastasis, yet understanding the intricate mechanisms and potential therapeutic targets remains nascent. Here it is reported that breast cancer establishes a hyperglycemic bone pre-metastatic niche before migrating to bone tissue and further enhances glucose metabolism following metastatic colonization. An intervention strategy is subsequently proposed targeting glucose metabolism utilizing a biomimetic-engineered enzyme-based nanoplatform. This platform's membrane shielding reduces the interaction between engineered glucose oxidase and circulating glucose, while the engineered enzyme specifically targets glucose metabolism, enabling self-amplifying starvation combined with selective chemotherapy. Such precision can precisely inhibit breast cancer bone metastases and block distal tumor dissemination. This study provides novel insights into the role of glucose metabolism in the pre-metastatic niche and presents a proof-of-concept for metabolic-targeted strategies in breast cancer bone metastasis treatment. This approach holds significant promise for improving therapeutic outcomes in metastatic breast cancer by targeting the metabolic vulnerabilities of the bone microenvironment and halting systemic tumor spread.
Collapse
Affiliation(s)
- Jianxin Ye
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Peng Hu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200080, China
| | - Rui Zhang
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Lei Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zonghua Luo
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, 200080, China
| | - Yanan Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200080, China
| | - Shengzhe Ruan
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Mengyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200080, China
| | - Huaze Ding
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yike Qian
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Tong Meng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Changping Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
4
|
Hu JJ, Zhang QY, Yang ZC. The correlation between obesity and the occurrence and development of breast cancer. Eur J Med Res 2025; 30:419. [PMID: 40414892 DOI: 10.1186/s40001-025-02659-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 05/04/2025] [Indexed: 05/27/2025] Open
Abstract
This study reviews the mechanisms by which obesity affects the development and progression of breast cancer (BC). The association between obesity and BC is mainly due to three aspects: disruption of glycolipid metabolism, abnormal cell function and imbalance of adipokine levels. The dysregulation of glycolipid metabolism caused by obesity, including the accumulation of cholesterol and fatty acids and the reprogramming of glucose metabolism, promotes the growth and invasion of tumour cells. Obesity triggers multiple cellular abnormalities, particularly in lipid-associated macrophages and cancer-associated adipocytes, which promote tumour progression and immunosuppression by secreting inflammatory factors and various fatty acids into the tumour microenvironment. Obesity leads to an imbalance in the expression of several adipokines. Leptin upregulation is closely associated with BC metastasis and resistance to endocrine therapy, while reduced adiponectin levels attenuate the protective effect. At the same time, chronic inflammation and insulin resistance not only further increase the risk of BC, but also exacerbate tumour resistance. In terms of treatment, weight-loss drugs and metformin can improve the efficacy of obesity-related BC treatment to some extent. Intervention strategies targeting adipose tissue remodelling, lipid metabolism and leptin regulation also show potential clinical value, but more research is needed to clarify their safety and efficacy. This review provides systematic ideas and references for research into the mechanisms and clinical management of obesity-related BC.
Collapse
Affiliation(s)
- Jun-Jie Hu
- Hunan University of Traditional Chinese Medicine, Changsha, 410078, China
| | - Qi-Yue Zhang
- Hunan University of Traditional Chinese Medicine, Changsha, 410078, China
| | - Zhi-Chun Yang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
5
|
Esposito T, Pentimalli F, Giordano A, Cortellino S. Vitamins and dietary supplements in cancer treatment: is there a need for increased usage? Expert Rev Anticancer Ther 2025:1-24. [PMID: 40322898 DOI: 10.1080/14737140.2025.2501077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Vitamins are essential for homeostasis and proper functioning of organisms. These micronutrients prevent tumor onset by functioning as antioxidants and enzymatic cofactors involved in anti-stress and immune responses, modulating epigenetic regulators, and shaping the microbiota composition. Unbalanced diets and sedentary lifestyles contribute to obesity, associated with increasing cancer risk. Cancer patients often exhibit vitamin deficiencies due to chronic inflammation, anticancer therapies, and tumor-induced metabolic changes, leading to malnutrition and cachexia. AREAS COVERED This review critically analyzes preclinical and clinical studies, sourced from PubMed and ClinicalTrials.gov databases, that investigate the potential benefits of vitamin supplementation and dietary interventions, such as intermittent fasting and ketogenic diets, in mouse tumor models and cancer patients. This analysis elucidates the limitations of such interventions and suggests optimal dietary strategies to prevent cancer and enhance patients' quality of life and prognosis. EXPERT OPINION To date, clinical studies have found no substantial benefit of over-the-counter vitamin supplements and dietary interventions on cancer patients' health and prognosis. To prevent the spread of useless and potentially harmful products by the nutraceutical industry, establishing a regulatory authority is necessary to monitor and ensure product quality and validity before commercialization.
Collapse
Affiliation(s)
- Teresa Esposito
- Department of Clinical Dietetics and Metabolic Diseases, Cavalier Raffaele Apicella Hospital, ASL Napoli 3 Sud, Naples, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro", Bari, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Salvatore Cortellino
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, Italy
- S.H.R.O. Italia Foundation ETS, Turin, Italy
| |
Collapse
|
6
|
Mohammadzadeh M, Amirpour M, Ahmadirad H, Abdi F, Khalesi S, Naghshi N, Bahrami A, Hejazi E. Impact of Fasting Mimicking Diet (FMD) on cardiovascular risk factors: a systematic review and meta-analysis of randomized control trials. Diabetol Metab Syndr 2025; 17:137. [PMID: 40287774 PMCID: PMC12032729 DOI: 10.1186/s13098-025-01709-5] [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: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The Fasting Mimicking Diet (FMD) has gained significant attention as a potential intervention for reducing cardiovascular risk factors. While studies have investigated its effectiveness, findings have been inconsistent. Therefore, this systematic review and meta-analysis aimed to clarify evidence on the impact of FMD on cardiovascular risk factors. METHOD PubMed, Web of Science, Scopus, and Google Scholar were searched for eligible Randomized controlled trials (RCTs) published up to July 2024. Weighted mean differences (WMD) were calculated for the net changes in risk factors using random-effects models. RESULTS Eleven RCTs (with twelve treatment arms) were included. FMD significantly reduced glycated hemoglobin (HbA1c) (WMD = -8.589 mmol/mol, 95% CI: -12.389, -4.769), insulin-like growth factor 1 (IGF-1) (WMD= -19.211 ng/ml, 95% CI: -32.986, -5.437), systolic blood pressure (SBP) (WMD = -4.148 mmHg, 95% CI: -7.584, -0.711), and diastolic blood pressure (DBP) (WMD = -2.263 mmHg, 95% CI: -4.162, -0.364) levels. No significant effects were observed on other cardiovascular risk factors. CONCLUSION This meta-analysis suggests that FMD can significantly reduce HbA1c, IGF-1, SBP, and DBP levels. Further research is warranted to investigate the long-term and potential clinical implications of FMD on cardiovascular health. PROSPERO REGISTRATION The protocol of the study was registered in the International Prospective Register of Systematic Reviews (PROSPERO registration no: CRD42024569426).
Collapse
Affiliation(s)
- Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Amirpour
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahmadirad
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abdi
- Department of Nutrition, Faculty of Nutrition and food science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saman Khalesi
- Appleton Institute, School of Health, Medical and Applied Sciences, Central Queensland University, Brisbane, Australia
| | - Niayesh Naghshi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Bahrami
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Hejazi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Nan K, Zhong Z, Yue Y, Shen Y, Zhang H, Wang Z, Zhuma K, Yu B, Fu Y, Wang L, Sun X, Qu M, Chen Z, Guo M, Zhang J, Chu Y, Liu R, Miao C. Fasting-mimicking diet-enriched Bifidobacterium pseudolongum suppresses colorectal cancer by inducing memory CD8 + T cells. Gut 2025; 74:775-786. [PMID: 39870395 DOI: 10.1136/gutjnl-2024-333020] [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: 06/03/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Fasting-mimicking diet (FMD) boosts the antitumour immune response in patients with colorectal cancer (CRC). The gut microbiota is a key host immunity regulator, affecting physiological homeostasis and disease pathogenesis. OBJECTIVE We aimed to investigate how FMD protects against CRC via gut microbiota modulation. DESIGN We assessed probiotic species enrichment in FMD-treated CRC mice using faecal metagenomic sequencing. The candidate species were verified in antibiotic-treated conventional and germ-free mouse models. Immune landscape alterations were evaluated using single-cell RNA sequencing and multicolour flow cytometry. The microbiota-derived antitumour metabolites were identified using metabolomic profiling. RESULTS Faecal metagenomic profiling revealed Bifidobacterium pseudolongum enrichment in FMD-treated CRC mice. B. pseudolongum mediates the FMD antitumour effects by increasing the tissue-resident memory CD8+ T-cell (TRM) population in CRC mice. The level of L-arginine, a B. pseudolongum functional metabolite, increased in FMD-treated CRC mice; furthermore, L-arginine induced the TRM phenotype in vivo and in vitro. Mechanistically, L-arginine is transported by the solute carrier family 7-member 1 (SLC7A1) receptor in CD8+ T cells. Both FMD and B. pseudolongum improved anti-CTLA-4 efficacy in the orthotopic mouse CRC model. In FMD-treated patients with CRC, the CD8+ TRM cell number increased as B. pseudolongum and L-arginine accumulated. The abundance of CD8+ TRM cells and B. pseudolongum was associated with a better prognosis in patients with CRC. CONCLUSION B. pseudolongum contributes to the FMD antitumour effects in CRC by producing L-arginine. This promotes CD8+ T-cell differentiation into memory cells. B. pseudolongum administration is a potential CRC therapeutic strategy.
Collapse
Affiliation(s)
- Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Zhiqiang Wang
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Kameina Zhuma
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ying Fu
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xingfeng Sun
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Anesthesiology, Obstetrics and Gynecology, Hospital of Fudan University, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Zhaoyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Miaomiao Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Jie Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, and Shanghai Clinical Research Center for Anesthesiology, Shanghai, China
| |
Collapse
|
8
|
Abene J, Deng J. Evaluating the role of dietary interventions in reducing chemotherapy toxicities in cancer patients: a systematic review. J Cancer Surviv 2025:10.1007/s11764-025-01777-6. [PMID: 40119985 DOI: 10.1007/s11764-025-01777-6] [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: 02/11/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE This systematic review synthesizes current literature examining the relationship between various dietary patterns and chemotherapy toxicities among patients currently receiving chemotherapy treatment. It aims to determine the most advantageous dietary pattern for patients with various malignancies. METHODS PubMed, CINAHL, and Embase were thoroughly searched to select quantitative studies that answered the research question and met the inclusion criteria as of July 2024. RESULTS Fourteen studies were analyzed: 10 randomized control trials (RCTs), 2 quasi-experimental, 1 case series, and 1 cohort study. Analyses were stratified by dietary pattern: fasting; Mediterranean diet; ketogenic diet; plant-based, high-protein diet; and anti-inflammatory diet. The results suggest that these all have the ability to benefit patients' experiences with chemotherapy. Fasting; a Mediterranean diet; a plant-based, high-protein diet; and an anti-inflammatory diet may reduce toxicities such as fatigue, diarrhea, insomnia, and nausea throughout chemotherapy. The ketogenic diet might offer initial advantages in improving diarrhea and physical activity; however, these results may not be sustainable. CONCLUSIONS There is insufficient evidence to identify a single dietary intervention as the most effective in reducing chemotherapy toxicities in patients undergoing chemotherapy. Further research is necessary to uncover the most beneficial dietary pattern to recommend as a nutritional strategy for survivors undergoing chemotherapy. IMPLICATIONS FOR CANCER SURVIVORS Survivors may benefit from adopting specific dietary patterns to alleviate chemotherapy toxicities. These reductions may improve chemotherapy tolerance, minimize treatment interruptions, and enhance daily functioning.
Collapse
Affiliation(s)
- Jessica Abene
- School of Nursing, University of Pennsylvania, 418 Curie Blvd, Philadelphia, PA, USA.
| | - Jie Deng
- School of Nursing, University of Pennsylvania, 418 Curie Blvd, Philadelphia, PA, USA
| |
Collapse
|
9
|
Liu X, Meng Q, Fan W, Ning L, Ge L. The effects of intermittent fasting on anthropometric indices, glycemic profile, chemotherapy-related toxicity, and subjective perception in gynecological and breast cancer patients: a systematic review and meta-analysis. BMC Cancer 2025; 25:419. [PMID: 40055608 PMCID: PMC11887389 DOI: 10.1186/s12885-025-13806-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Mounting evidence supports the health benefits of intermittent fasting (IF) in general. This study evaluates its impact on patients with gynecological or breast cancer specifically. METHODS A thorough search for studies comparing IF with either nonintervention diets or calorie restriction (CR) in patients with either gynecological or breast cancer and published prior to October 5, 2024 was carried out on the PubMed, Web of Science, Cochrane Library, Scopus, Embase, China National Knowledge Infrastructure (CNKI), and Chinese Biomedical Literature databases (CBM). Extracted data included but not limited to body mass index (BMI), body weight, waist circumference (WC), fasting glucose, insulin levels, chemotherapy-related toxicity, and subjective perceptions. RESULTS A total of 625 subjects were included across 7 randomized controlled trials, and 2 nonrandomized trials. Meta-analysis revealed that IF significantly reduced body weight (Effect Size [ES]: -0.611; 95% Confidence Interval [CI]: -0.886 to -0.356; p < 0.001; I² = 0%), blood glucose levels (standardized mean difference [SMD]: -0.347 mmol/L; 95% CI: -0.533 to -0.140; p < 0.001), and insulin concentrations (SMD: -0.395 mU/L; 95% CI: -0.674 to -0.116; p = 0.005). Sensitivity analysis indicated that the overall effect sizes were stable. However, it remains uncertain whether IF increases chemotherapy-related adverse effects (relative risk [RR]: 1.038; 95% CI: 0.844 to 1.278; p = 0.723). Furthermore, three studies indicated that IF reduced fatigue and two studies indicated that IF improved quality of life. CONCLUSION This systematic review and meta-analysis suggests that IF has a beneficial effect on reducing body weight, blood glucose, and insulin concentrations in gynecological and breast cancer patients. IF may also reduce fatigue and improve quality of life. However, the effect on chemotherapy-related adverse effects is uncertain. Further high-quality studies with long-term follow-ups are needed to confirm these findings.
Collapse
Affiliation(s)
- Xiaoxia Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Qiucen Meng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Wenqi Fan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Lianzhen Ning
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Lina Ge
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
10
|
Chakos K, Pezley L, Bryner A, Czyzewski O, Corral J, Marquina V, Gadi V, Ganschow P, Tussing-Humphreys L, Gabel K. Impact of Diet Modifications on Body Weight, Body Composition, Treatment Outcomes, and Quality of Life During Primary Treatment for Breast Cancer: A Systematic Review. Nutr Rev 2025; 83:479-502. [PMID: 39190892 PMCID: PMC11819483 DOI: 10.1093/nutrit/nuae109] [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: 08/29/2024] Open
Abstract
CONTEXT Breast cancer is a significant public health challenge, with 290 000 new cases annually and significant healthcare costs. Treatment advancements have led to improvements in survival, but common adverse effects include weight gain, fatigue, nausea, and taste changes, decreasing quality of life. OBJECTIVE This review aims to assess the impact of diet and lifestyle interventions during primary treatment for breast cancer and their effects on body weight, body composition, treatment-related adverse outcomes, and patient-reported quality of life. DATA SOURCES AND DATA EXTRACTION A search of PubMed, CINAHL, and EMBASE conducted through May 10, 2023, identified 31 publications describing 27 interventions including diet or diet plus exercise. The Cochrane Risk of Bias tool assessed the quality of publications. DATA ANALYSIS The findings suggest that whole foods, aerobic and strength-training exercises, and intermittent fasting during treatment may improve body weight and composition, treatment-related outcomes, and quality of life. Limitations include variation in study duration, small sample sizes, and limited sociodemographic data. CONCLUSION Improvements seen with increased diet quality and reduced caloric intake, with or without exercise, challenge current standard-of-care recommendations during treatment for breast cancer. While there is a need for additional research, healthcare teams can confidently promote healthy diets and exercise during primary treatment for breast cancer to manage weight and improve treatment-related side effects and quality of life. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. 42023425613.
Collapse
Affiliation(s)
- Kaitlin Chakos
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Lacey Pezley
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Alyssa Bryner
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Ola Czyzewski
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Jennifer Corral
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
- University of Illinois Cancer Center, Chicago, IL 60612, United States
| | - Vanessa Marquina
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Vijayakrishna Gadi
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
- University of Illinois Cancer Center, Chicago, IL 60612, United States
| | - Pamela Ganschow
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
- University of Illinois Cancer Center, Chicago, IL 60612, United States
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
- University of Illinois Cancer Center, Chicago, IL 60612, United States
| | - Kelsey Gabel
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, United States
| |
Collapse
|
11
|
Fatima G, Mehdi AA, Fedacko J, Hadi N, Magomedova A, Mehdi A. Fasting as Cancer Treatment: Myth or Breakthrough in Oncology. Cureus 2025; 17:e81395. [PMID: 40296920 PMCID: PMC12035504 DOI: 10.7759/cureus.81395] [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: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The concept of fasting as a potential cancer treatment has garnered increasing interest, particularly in light of emerging evidence linking dietary interventions to cancer progression and therapy outcomes. This article explores whether fasting, either intermittent or prolonged, can be a viable standalone treatment for cancer or if its therapeutic potential lies in its adjunctive role. Current research suggests that fasting induces a metabolic shift, which may inhibit cancer cell proliferation by depriving them of essential nutrients. Additionally, fasting has been shown to enhance the body's stress resistance, promote autophagy, and possibly make cancer cells more vulnerable to standard treatments such as chemotherapy and radiotherapy. However, the application of fasting as a sole treatment for cancer remains controversial and lacks substantial clinical validation. While animal models and in vitro studies indicate promising results, the translation to human trials is complex, with various types of cancer responding differently to dietary interventions. Moreover, concerns about malnutrition, loss of muscle mass, and the overall health of cancer patients undergoing fasting without supervision must be addressed. The paper critically examines the myth and reality surrounding fasting as a cancer treatment, reviewing key studies and clinical trials to provide a comprehensive understanding of its efficacy and safety. While fasting may hold promise as a supportive therapy, particularly in combination with traditional treatments, there is currently insufficient evidence to support its use as a primary treatment modality. Further research is needed to establish the parameters in which fasting might be beneficial, such as specific cancer types, patient populations, and optimal fasting regimens. Thus, while the idea of fasting as a cancer breakthrough is compelling, it remains a complementary approach rather than a standalone solution in oncology.
Collapse
Affiliation(s)
- Ghizal Fatima
- Department of Public Health, Era's Lucknow Medical College and Hospital, Era University, Lucknow, IND
| | - Abbas A Mehdi
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, IND
| | - Jan Fedacko
- Department of Cardiology, Pavol Jozef Safarik University, Kosice, Kosice, SVK
| | - Najah Hadi
- Department of Medicine, University of Kufa, Najaf, IRQ
| | | | - Ammar Mehdi
- Department of Pediatric Dentistry, Career Post Graduate Institute of Dental Sciences and Hospital, Lucknow, IND
| |
Collapse
|
12
|
Li R, Ma Y, He A, Pu Y, Wan X, Sun H, Wang N, Luo M, Wang G, Xia Y. Fasting enhances the efficacy of Sorafenib in breast cancer via mitophagy mediated ROS-driven p53 pathway. Free Radic Biol Med 2025; 229:350-363. [PMID: 39864757 DOI: 10.1016/j.freeradbiomed.2025.01.047] [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: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
The multi-kinase inhibitor sorafenib has shown potential to inhibit tumor cell growth and intra-tumoral angiogenesis by targeting several kinases, including VEGFR2 and RAF. Abnormal activation of the Ras/Raf/MAPK/ERK kinase cascade and the VEGF pathway is a common feature in breast cancer. However, the efficacy of sorafenib in breast cancer treatment remains limited. Recently, fasting has emerged as a promising non-pharmacological approach to modulate cancer metabolism and enhance the effectiveness of cancer therapies. In this study, we found that fasting significantly enhances the anti-cancer effects of sorafenib monotherapy and its combination with immunotherapy in breast cancer models without causing obvious side effects. This combined treatment effectively inhibits tumor cell proliferation and intra-tumoral angiogenesis. The fasting-induced reduction in peripheral blood glucose levels strongly correlated with enhanced sensitivity to sorafenib. Mechanistically, the combined treatment induced mitophagy, characterized by mitochondrial dysfunction and activation of the PINK1-Parkin pathway. Consequently, increased mitochondrial ROS levels promoted p53 expression, amplifying cell cycle arrest and apoptosis in breast cancer cells. Furthermore, fasting reduced lactate levels within the tumor, and the consequent glucose limitation synergized with sorafenib to activate AMPK, which in turn elevated PD-L1 expression in tumor cells, potentially enhancing their sensitivity to immunotherapy. In summary, our findings demonstrate that fasting and sorafenib, as a rational combination therapy, induce mitophagy, thereby enhancing sorafenib's efficacy in treating breast cancer through the ROS-driven p53 pathway. This study underscores the potential of fasting in breast cancer therapy and provides a foundation for optimizing the clinical application of sorafenib.
Collapse
Affiliation(s)
- Ru Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China
| | - Yimei Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Anqi He
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yamin Pu
- Department of Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuanting Wan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Hongbao Sun
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Min Luo
- Department of Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China.
| | - Yong Xia
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Mondino A, Baù MG, Surace A, Gallo F, Bortolini M, Mano MP. Fasting, Physical Activity, and Art Therapy Improve Metabolic Syndrome and Inflammasomes: An Italian Residential Experience. J Lifestyle Med 2025; 15:1-7. [PMID: 40376694 PMCID: PMC12076049 DOI: 10.15280/jlm.2025.15.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/18/2024] [Accepted: 02/19/2025] [Indexed: 05/18/2025] Open
Abstract
Background Diet, physical activity, and general lifestyle modifications are implicated in metabolic pathway prevention and may improve some chronic diseases such as metabolic syndrome. The aim of this study is to measure the effectiveness of a short-time nutrition restriction regime combined with a daily practice of physical and creative activities on metabolic syndrome parameters, inflammasome status, and the compliance of the patients to the proposed scheme. Methods This pilot study analyzed 95 volunteers who took part in a residential one-week lasting experimental lifestyle period. The program proposed was structured in a four-day detox nutrition based only on vegetable products derived from organic, biologic, synergic, and integrated farming, without animal proteins, fats, and simple sugars. These four days were followed by a three-day fasting or fasting-mimicking diet, consisting of one or two hypocaloric meals based on complex sugars, vegetables, oleaginous seeds, and only in some cases vegetable proteins. Finally, a three-day "weaning" period was administered. During the whole period, 95 participants were offered a daily postural physical activity and a creative activity program. The basal data of the participants were collected (T0). A medical evaluation was conducted to collect the personal and anthropometric data. During this basal evaluation (T0), height, waist circumference, waist-to-height ratio (WtHR), glycemic value, and blood pressure were collected and reevaluated at the end of the 10-day experience (T1). Results The data were evaluated according to the three metabolic syndrome parameters (i.e., WtHR, glycemic value, and blood pressure). A significant improvement in the variables analyzed at the end of the program was observed. Conclusion This study revealed that a short integrated lifestyle program can influence some well-defined metabolic parameters and might decrease damage from metabolic syndrome.
Collapse
Affiliation(s)
- Aurelia Mondino
- Department of Gynecology and Obstetrics, ASL Città di Torino, Study Coordinator, Maria Vittoria Hospital, Torino, Italy
| | - Maria Grazia Baù
- Department of Gynecology and Obstetrics, Maria Vittoria Hospital, Torino, Italy
| | - Alessandra Surace
- Department of Gynecology and Obstetrics, Michele e Pietro Ferrero Hospital, Verduno, Italy
| | - Federica Gallo
- Department of Gynecology and Obstetrics, Epidemiology Unit, Health Direction, Local Health Authority of Cuneo 1, Cuneo, Italy
| | | | - Maria Piera Mano
- Department of Gynecology and Obstetrics, University of Turin, Sant'Anna Hospital, Torino, Italy
| |
Collapse
|
14
|
Cote AL, Munger CJ, Ringel AE. Emerging insights into the impact of systemic metabolic changes on tumor-immune interactions. Cell Rep 2025; 44:115234. [PMID: 39862435 DOI: 10.1016/j.celrep.2025.115234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/24/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Tumors are inherently embedded in systemic physiology, which contributes metabolites, signaling molecules, and immune cells to the tumor microenvironment. As a result, any systemic change to host metabolism can impact tumor progression and response to therapy. In this review, we explore how factors that affect metabolic health, such as diet, obesity, and exercise, influence the interplay between cancer and immune cells that reside within tumors. We also examine how metabolic diseases influence cancer progression, metastasis, and treatment. Finally, we consider how metabolic interventions can be deployed to improve immunotherapy. The overall goal is to highlight how metabolic heterogeneity in the human population shapes the immune response to cancer.
Collapse
Affiliation(s)
- Andrea L Cote
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Chad J Munger
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Alison E Ringel
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA.
| |
Collapse
|
15
|
Oudmaijer CAJ, Komninos DSJ, Ozinga RA, Smit K, Rozendaal NEM, Hoeijmakers JHJ, Vermeij WP, Aerts JGJV, IJzermans JNM, Willemsen M. Short-term fasting before living kidney donation has an immune-modulatory effect. Front Immunol 2025; 16:1488324. [PMID: 40051619 PMCID: PMC11882433 DOI: 10.3389/fimmu.2025.1488324] [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: 08/29/2024] [Accepted: 01/13/2025] [Indexed: 03/09/2025] Open
Abstract
Background Short-Term Fasting (STF) is an intervention reducing the intake of calories, without causing undernutrition or micronutrient-related malnutrition. It aims to systemically improve resilience against acute stress. Several (pre-)clinical studies have suggested protective effects of STF, marking the systemic effects STF can induce in respect to surgery and ischemia-reperfusion injury. In addition, STF also affects the number of circulating immune cells. We aim to determine the effect of STF on the abundance and phenotype of different immune cell populations. Methods Thirty participants were randomly selected from the FAST clinical trial, including living kidney donors, randomized to an STF-diet or control arm. In an observational cohort sub-study we prospectively included 30 patients who donated blood samples repeatedly during study runtime. Using flow cytometry analyses, immune cell phenotyping was performed on peripheral blood mononuclear cells. Three panels were designed to investigate the presence and activation status of peripheral T cells, B cells, dendritic cells (DCs) and myeloid cells. Results Eight participants were excluded due to sample constraints. Baseline characteristics showed no significant differences, except for fasting duration. Weight changes were minimal and non-significant across different time intervals, with slight trends toward long-term weight loss pre-surgery. Glucose, insulin, and β-hydroxybutyrate levels differed significantly between groups, reflecting adherence to the fasting diet. Flow cytometry and RNA sequencing analysis revealed no baseline differences between groups, with high variability within each group. STF changes the levels and phenotype of immune cells, reducing the abundance and activation of T cells, including regulatory T cells, increased presence of (naïve) B cells, and elevation of type 1 conventional DCs (cDC1s). In addition, a decrease in central memory T cells was observed. Discussion In this study, we observed significant changes due to fasting in B cells, T cells, and DCs, specifically toward less specialized lymphocytes, suggesting an arrest in B and T cell development. Further research should focus on the clinical implications of changes in immune cells and significance of these observed immunological changes. Conclusion STF results in reduced numbers and activation status of T cells and Tregs, increased presence of (naïve) B cells, and elevation of cDC1s.
Collapse
Affiliation(s)
- Christiaan A. J. Oudmaijer
- Erasmus MC Transplant Institute, Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Daphne S. J. Komninos
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Rutger A. Ozinga
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Kimberly Smit
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Nina E. M. Rozendaal
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jan H. J. Hoeijmakers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Erasmus MC Cancer Institute, Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Germany, and Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Wilbert P. Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Joachim G. J. V. Aerts
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jan N. M. IJzermans
- Erasmus MC Transplant Institute, Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marcella Willemsen
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
16
|
Micarelli A, Mrakic-Sposta S, Vezzoli A, Malacrida S, Caputo S, Micarelli B, Misici I, Carbini V, Iennaco I, Granito I, Longo VD, Alessandrini M. Chemosensory and cardiometabolic improvements after a fasting-mimicking diet: A randomized cross-over clinical trial. Cell Rep Med 2025; 6:101971. [PMID: 39970875 PMCID: PMC11866515 DOI: 10.1016/j.xcrm.2025.101971] [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: 05/08/2024] [Revised: 09/26/2024] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Obesity is associated with a decrease in chemosensory perception acuity and increased disease risk, pointing to the need for feasible interventions that affect smell, taste, and cardiometabolic markers. Here, subjects with overweight/obesity are treated with six monthly cycles of a fasting-mimicking diet (FMD) lasting 5 days followed by a normal diet for the rest of the month to determine their effects on chemosensory function and cardiometabolic risk factors. Both arms of the 102 randomized cross-over trial participants indicate FMD-dependent improvements in a wide range of taste and smell chemosensory functions. The portion of hyposmic subjects is reduced from 38.1% at baseline to 6.4% at the end of 6 FMD cycles. FMD cycles also reduce cardiometabolic and inflammatory markers and drug use in diabetic patients. This trial provides evidence for the effect of periodic FMD cycles in improving chemosensory function while reducing cardiometabolic risk factors without requiring long-term lifestyle changes. The trial is registered at ClinicalTrials.gov (NCT04529161).
Collapse
Affiliation(s)
- Alessandro Micarelli
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy.
| | | | - Alessandra Vezzoli
- Institute of Clinical Physiology, National Research Council (CNR), Milan, Italy
| | - Sandro Malacrida
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
| | | | - Beatrice Micarelli
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Ilaria Misici
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Valentina Carbini
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Ilaria Iennaco
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Ivan Granito
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| | - Marco Alessandrini
- University of Rome Tor Vergata - Department of Clinical Sciences and Translational Medicine - Ear-Nose-Throat Unit, Rome, Italy
| |
Collapse
|
17
|
Ligorio F, Vingiani A, Torelli T, Sposetti C, Drufuca L, Iannelli F, Zanenga L, Depretto C, Folli S, Scaperrotta G, Capri G, Bianchi GV, Ferraris C, Martelli G, Maugeri I, Provenzano L, Nichetti F, Agnelli L, Lobefaro R, Fucà G, Fotia G, Mariani L, Morelli D, Ladisa V, De Santis MC, Lozza L, Trecate G, Belfiore A, Brich S, Bertolotti A, Lorenzini D, Ficchì A, Martinetti A, Sottotetti E, Arata A, Corsetto P, Sorrentino L, Rediti M, Salvadori G, Minucci S, Foiani M, Apolone G, Pagani M, Pruneri G, de Braud F, Vernieri C. Early downmodulation of tumor glycolysis predicts response to fasting-mimicking diet in triple-negative breast cancer patients. Cell Metab 2025; 37:330-344.e7. [PMID: 39694040 DOI: 10.1016/j.cmet.2024.11.004] [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: 07/24/2024] [Revised: 09/04/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024]
Abstract
In preclinical experiments, cyclic fasting-mimicking diets (FMDs) showed broad anticancer effects in combination with chemotherapy. Among different tumor types, triple-negative breast cancer (TNBC) is exquisitely sensitive to FMD. However, the antitumor activity and efficacy of cyclic FMD in TNBC patients remain unclear. Here, we show that a severely calorie-restricted, triweekly, 5-day FMD regimen results in excellent pathologic complete response (pCR) rates (primary endpoint) and long-term clinical outcomes (secondary endpoints) when combined with preoperative chemotherapy in 30 patients with early-stage TNBC enrolled in the phase 2 trial BREAKFAST. Bulk and single-cell RNA sequencing analysis revealed that highly glycolytic cancer cells, myeloid cells, and pericytes from tumors achieving pCR undergo a significant, early downmodulation of pathways related to glycolysis and pyruvate metabolism. Our findings pave the wave for conducting larger clinical trials to investigate the efficacy of cyclic FMD in early-stage TNBC patients and to validate early changes of intratumor glycolysis as a predictor of clinical benefit from nutrient restriction. This study was registered at Clinicaltrials.gov (NCT04248998).
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Andrea Vingiani
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Tommaso Torelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Caterina Sposetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Lorenzo Drufuca
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Fabio Iannelli
- Haematopathogy Division, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Lucrezia Zanenga
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Catherine Depretto
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Secondo Folli
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Gianfranco Scaperrotta
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giuseppe Capri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giulia V Bianchi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Cristina Ferraris
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Gabriele Martelli
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Ilaria Maugeri
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Federico Nichetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Luca Agnelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giuseppe Fotia
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Luigi Mariani
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Daniele Morelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Vito Ladisa
- Hospital Pharmacy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Maria Carmen De Santis
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Laura Lozza
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giovanna Trecate
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Antonino Belfiore
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Silvia Brich
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Alessia Bertolotti
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Daniele Lorenzini
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Angela Ficchì
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Alessio Arata
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Paola Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Luca Sorrentino
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Mattia Rediti
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giulia Salvadori
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Saverio Minucci
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Marco Foiani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Massimiliano Pagani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Giancarlo Pruneri
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.
| |
Collapse
|
18
|
López-Cánovas JL, Naranjo-Martínez B, Diaz-Ruiz A. Fasting in combination with the cocktail Sorafenib:Metformin blunts cellular plasticity and promotes liver cancer cell death via poly-metabolic exhaustion. Cell Oncol (Dordr) 2025; 48:161-182. [PMID: 38990489 PMCID: PMC11850423 DOI: 10.1007/s13402-024-00966-2] [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] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
PURPOSE Dual-Interventions targeting glucose and oxidative metabolism are receiving increasing attention in cancer therapy. Sorafenib (S) and Metformin (M), two gold-standards in liver cancer, are known for their mitochondrial inhibitory capacity. Fasting, a glucose-limiting strategy, is also emerging as chemotherapy adjuvant. Herein, we explore the anti-carcinogenic response of nutrient restriction in combination with sorafenib:metformin (NR-S:M). RESULTS Our data demonstrates that, independently of liver cancer aggressiveness, fasting synergistically boosts the anti-proliferative effects of S:M co-treatment. Metabolic and Cellular plasticity was determined by the examination of mitochondrial and glycolytic activity, cell cycle modulation, activation of cellular apoptosis, and regulation of key signaling and metabolic enzymes. Under NR-S:M conditions, early apoptotic events and the pro-apoptotic Bcl-xS/Bcl-xL ratio were found increased. NR-S:M induced the highest retention in cellular SubG1 phase, consistent with the presence of DNA fragments from cellular apoptosis. Mitochondrial functionality, Mitochondrial ATP-linked respiration, Maximal respiration and Spare respiratory capacity, were all found blunted under NR-S:M conditions. Basal Glycolysis, Glycolytic reserve, and glycolytic capacity, together with the expression of glycogenic (PKM), gluconeogenic (PCK1 and G6PC3), and glycogenolytic enzymes (PYGL, PGM1, and G6PC3), were also negatively impacted by NR-S:M. Lastly, a TMT-proteomic approach corroborated the synchronization of liver cancer metabolic reprogramming with the activation of molecular pathways to drive a quiescent-like status of energetic-collapse and cellular death. CONCLUSION Altogether, we show that the energy-based polytherapy NR-S:M blunts cellular, metabolic and molecular plasticity of liver cancer. Notwithstanding the in vitro design of this study, it holds a promising therapeutic tool worthy of exploration for this tumor pathology.
Collapse
Affiliation(s)
- Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Alberto Diaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, Spain.
| |
Collapse
|
19
|
Wang R, Lv X, Xu W, Li X, Tang X, Huang H, Yang M, Ma S, Wang N, Niu Y. Effects of the periodic fasting-mimicking diet on health, lifespan, and multiple diseases: a narrative review and clinical implications. Nutr Rev 2025; 83:e412-e426. [PMID: 38287649 DOI: 10.1093/nutrit/nuae003] [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: 01/31/2024] Open
Abstract
Dietary restriction and fasting have been recognized for their beneficial effects on health and lifespan and their potential application in managing chronic metabolic diseases. However, long-term adherence to strict dietary restrictions and prolonged fasting poses challenges for most individuals and may lead to unhealthy rebound eating habits, negatively affecting overall health. As a result, a periodic fasting-mimicking diet (PFMD), involving cycles of fasting for 2 or more days while ensuring basic nutritional needs are met within a restricted caloric intake, has gained widespread acceptance. Current research indicates that a PFMD can promote stem cell regeneration, suppress inflammation, extend the health span of rodents, and improve metabolic health, among other effects. In various disease populations such as patients with diabetes, cancer, multiple sclerosis, and Alzheimer's disease, a PFMD has shown efficacy in alleviating disease symptoms and improving relevant markers. After conducting an extensive analysis of available research on the PFMD, it is evident that its advantages and potential applications are comparable to other fasting methods. Consequently, it is proposed in this review that a PFMD has the potential to fully replace water-only or very-low-energy fasting regimens and holds promise for application across multiple diseases.
Collapse
Affiliation(s)
- Ruohua Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Xinyi Lv
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Wenyu Xu
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Xiaoqing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Xuanfeng Tang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - He Huang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Mengxia Yang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Shuran Ma
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Nan Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Yucun Niu
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| |
Collapse
|
20
|
Silvestris N, Aprile G, Tessitore D, Mentrasti G, Cristina Petrella M, Speranza D, Casirati A, Caccialanza R, Cinieri S, Pedrazzoli P. Harnessing tumor metabolism during cancer treatment: A narrative review of emerging dietary approaches. Crit Rev Oncol Hematol 2025; 206:104571. [PMID: 39581244 DOI: 10.1016/j.critrevonc.2024.104571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024] Open
Abstract
Cancer is currently one of the biggest public health challenges worldwide, ranking as the second leading cause of death globally. To date, strong epidemiological associations have been demonstrated between unhealthy lifestyles and eating habits, i.e. obesity, and an increased risk of developing cancer. However, there is limited evidence regarding the impact of specific dietary regimes on cancer outcomes during conventional cancer treatments. This paper systematically reviews and evaluates preclinical and clinical evidence regarding the effects of fasting, fast-mimicking diet, ketogenic diet, vegan diet, alkaline diet, paleolithic diet, the Gerson regimen, and macrobiotic diet in the context of cancer treatments. Clinical trials on dietary regimes as complementary cancer therapy are limited by significant differences in trial design, patient characteristics, and cancer type, making it difficult to draw conclusions. In the future, more uniformly controlled clinical trials should help to better define the role of diets in cancer management.
Collapse
Affiliation(s)
- Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Dalila Tessitore
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| | - Giulia Mentrasti
- Medical Oncology, University Hospital-Marche Polytechnic University, Ancona, Italy
| | | | - Desirèe Speranza
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| | - Amanda Casirati
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy.
| | - Paolo Pedrazzoli
- Department of Internal Medicine, University of Pavia, Pavia, Italy; Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
21
|
Caprara G, Pallavi R, Sanyal S, Pelicci PG. Dietary Restrictions and Cancer Prevention: State of the Art. Nutrients 2025; 17:503. [PMID: 39940361 PMCID: PMC11820753 DOI: 10.3390/nu17030503] [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: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Worldwide, almost 10 million cancer deaths occurred in 2022, a number that is expected to rise to 16.3 million by 2040. Primary prevention has long been acknowledged as a crucial approach to reducing cancer incidence. In fact, between 30 and 50 percent of all tumors are known to be preventable by eating a healthy diet, staying active, avoiding alcohol, smoking, and being overweight. Accordingly, many international organizations have created tumor prevention guidelines, which underlie the importance of following a diet that emphasizes eating plant-based foods while minimizing the consumption of red/processed meat, sugars, processed foods, and alcohol. However, further research is needed to define the relationship between the effect of specific diets or nutritional components on cancer prevention. Interestingly, reductions in food intake and dietetic restrictions can extend the lifespan of yeast, nematodes, flies, and rodents. Despite controversial results in humans, those approaches have the potential to ameliorate health via direct and indirect effects on specific signaling pathways involved in cancer onset. Here, we describe the latest knowledge on the cancer-preventive potential of dietary restrictions and the biochemical processes involved. Molecular, preclinical, and clinical studies evaluating the effects of different fasting strategies will also be reviewed.
Collapse
Affiliation(s)
- Greta Caprara
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Shalini Sanyal
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| |
Collapse
|
22
|
Zuo Q, Kang Y. Metabolic Reprogramming and Adaption in Breast Cancer Progression and Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:347-370. [PMID: 39821033 DOI: 10.1007/978-3-031-70875-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Recent evidence has revealed that cancer is not solely driven by genetic abnormalities but also by significant metabolic dysregulation. Cancer cells exhibit altered metabolic demands and rewiring of cellular metabolism to sustain their malignant characteristics. Metabolic reprogramming has emerged as a hallmark of cancer, playing a complex role in breast cancer initiation, progression, and metastasis. The different molecular subtypes of breast cancer exhibit distinct metabolic genotypes and phenotypes, offering opportunities for subtype-specific therapeutic approaches. Cancer-associated metabolic phenotypes encompass dysregulated nutrient uptake, opportunistic nutrient acquisition strategies, altered utilization of glycolysis and TCA cycle intermediates, increased nitrogen demand, metabolite-driven gene regulation, and metabolic interactions with the microenvironment. The tumor microenvironment, consisting of stromal cells, immune cells, blood vessels, and extracellular matrix components, influences metabolic adaptations through modulating nutrient availability, oxygen levels, and signaling pathways. Metastasis, the process of cancer spread, involves intricate steps that present unique metabolic challenges at each stage. Successful metastasis requires cancer cells to navigate varying nutrient and oxygen availability, endure oxidative stress, and adapt their metabolic processes accordingly. The metabolic reprogramming observed in breast cancer is regulated by oncogenes, tumor suppressor genes, and signaling pathways that integrate cellular signaling with metabolic processes. Understanding the metabolic adaptations associated with metastasis holds promise for identifying therapeutic targets to disrupt the metastatic process and improve patient outcomes. This chapter explores the metabolic alterations linked to breast cancer metastasis and highlights the potential for targeted interventions in this context.
Collapse
Affiliation(s)
- Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
23
|
Li N, Sun YJ, Huang LY, Li RR, Zhang JS, Qiu AH, Wang J, Yang L. Fasting-mimicking diet potentiates anti-tumor effects of CDK4/6 inhibitors against breast cancer by suppressing NRAS- and IGF1-mediated mTORC1 signaling. Drug Resist Updat 2025; 78:101161. [PMID: 39499997 DOI: 10.1016/j.drup.2024.101161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/13/2024] [Indexed: 12/18/2024]
Abstract
AIMS Acquired resistance to cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) frequently emerges, and CDK4/6i-containing therapies in triple-negative breast cancer (TNBC) remain to be determined. METHODS RNA-sequencing, cell viability analysis, immunoblotting, siRNA transfection et al. were used to investigate and verify the resistance mechanism. BALB/c nude mice xenograft models and spontaneous MMTV-PyMT models were used to explore in vivo efficacy. RESULTS The mTOR pathway was activated in acquired CDK4/6i-resistant cells and inhibition of mTORC1 restored the sensitivity. While fasting-mimicking diet (FMD) enhances the activity of anticancer agents by inhibiting the mTORC1 signaling, we assessed FMD and found that FMD restored the sensitivity of CDK4/6i-resistant cells to abemaciclib and potentiated the anti-tumor activity of CDK4/6i in TNBC. The anti-tumor effects of FMD and/or CDK4/6i were accompanied by the downregulation of S6 phosphorylation. FMD cooperated with CDK4/6i to suppress the levels of IGF1 and RAS. The combination of FMD and abemaciclib also led to a potent inhibition of tumor growth in spontaneous transgenic MMTV-PyMT mouse models. CONCLUSIONS Our data demonstrate that FMD overcomes resistance and potentiates the anti-tumor effect of CDK4/6i by inhibiting mTORC1 signaling via lowering the levels of IGF1 and RAS, providing the rationale for clinical investigation of a potential FMD-CDK4/6i strategy in breast cancer.
Collapse
Affiliation(s)
- Ning Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ya-Jie Sun
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Li-Yun Huang
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Rong-Rong Li
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Shantou University Medical College, Shantou University, Shantou, Guangdong 515000, China
| | - Jun-Sheng Zhang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ai-Hua Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jing Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Shantou University Medical College, Shantou University, Shantou, Guangdong 515000, China.
| |
Collapse
|
24
|
Habka D, Hsu WC, Antoun J. Economic Evaluation of Fasting Mimicking Diet Versus Standard Care in Diabetic Patients on Dual or Triple Medications at Baseline in the United States: A Cost-Utility Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:51-59. [PMID: 39343090 DOI: 10.1016/j.jval.2024.08.003] [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: 03/17/2024] [Revised: 07/24/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES According to most guidelines, dietary interventions are essential in the management of diabetes. Fasting has emerged as potential therapeutic regimes for diabetes. The proof-of-concept study and the fasting in diabetes treatment trial are the first to explore the clinical impact of the Fasting Mimicking Diet (FMD) in patients with type 2 diabetes mellitus. Their results showed that FMD cycles improve glycemic management and can be integrated into usual care complementary to current guidelines. This economic evaluation aims to assess the 10-year quality-of-life effects, cost implications, and cost-effectiveness of adding a 3-year FMD program to diabetes standard care in diabetic population on dual or triple medications at baseline from the perspective of the US payer. METHODS We constructed a microsimulation model in TreeAge using a published US-specific diabetes model. The model was populated using FMD effectiveness outcomes and publicly available clinical and economic data associated with diabetes complications, use of diabetes medications, hypoglycemia incidence, direct medical costs in 2021 USD, quality of life, and mortality. All benefits were discounted by 3%. RESULTS This cost-utility analysis showed that the FMD program was associated with 11.4% less diabetes complications, 67.2% less overall diabetes medication use, and 45.0% less hypoglycemia events over the 10-year simulation period. The program generated an additional effectiveness benefit of 0.211 quality-adjusted life year and net monetary benefit of 41 613 USD per simulated patient. Thus, the FMD program is cost saving. CONCLUSIONS These results indicate that the FMD program is a beneficial first-line strategy in T2DM management.
Collapse
|
25
|
Bornes L, van Winden LJ, Geurts VCM, de Bruijn B, Azarang L, Lanfermeijer M, Caruso M, Proost N, Boeije M, Lohuis JO, Belthier G, Noguera Delgado E, de Gruil N, Kroep JR, van de Ven M, Menezes R, Wesseling J, Kok M, Linn S, Broeks A, van Rossum HH, Scheele CLGJ, van Rheenen J. The oestrous cycle stage affects mammary tumour sensitivity to chemotherapy. Nature 2025; 637:195-204. [PMID: 39633046 PMCID: PMC11666466 DOI: 10.1038/s41586-024-08276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
The response of breast cancer to neoadjuvant chemotherapy (NAC) varies substantially, even when tumours belong to the same molecular or histological subtype1. Here we identify the oestrous cycle as an important contributor to this heterogeneity. In three mouse models of breast cancer, we show reduced responses to NAC when treatment is initiated during the dioestrus stage, when compared with initiation during the oestrus stage. Similar findings were observed in retrospective premenopausal cohorts of human patients. Mechanistically, the dioestrus stage exhibits systemic and localized changes, including (1) an increased number of cells undergoing epithelial-to-mesenchymal transition linked to chemoresistance2-4 and (2) decreased tumour vessel diameter, suggesting potential constraints to drug sensitivity and delivery. In addition, an elevated presence of macrophages, previously associated with chemoresistance induction5, characterizes the dioestrus phase. Whereas NAC disrupts the oestrous cycle, this elevated macrophage prevalence persists and depletion of macrophages mitigates the reduced therapy response observed when initiating treatment during dioestrus. Our data collectively demonstrate the oestrous cycle as a crucial infradian rhythm determining chemosensitivity, warranting future clinical studies to exploit optimal treatment initiation timing for enhanced chemotherapy outcomes.
Collapse
Affiliation(s)
- Laura Bornes
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lennart J van Winden
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Veerle C M Geurts
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Beaunelle de Bruijn
- VIB Center for Cancer Biology, KU Leuven Department of Oncology, Leuven, Belgium
| | - Leyla Azarang
- Biostatistics Centre & Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mirthe Lanfermeijer
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marika Caruso
- VIB Center for Cancer Biology, KU Leuven Department of Oncology, Leuven, Belgium
| | - Natalie Proost
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Manon Boeije
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jeroen O Lohuis
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Guillaume Belthier
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eulàlia Noguera Delgado
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nadia de Gruil
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renee Menezes
- Biostatistics Centre & Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sabine Linn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Huub H van Rossum
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
26
|
Abene J, Tyburski S, Kral TVE, Quinn R, Deng J. Diet as an Adjunct Therapy in Reducing Chemotherapy Toxicities and Improving Patients Quality of Life: A Systematic Review and Meta-Analysis. Nutr Cancer 2024; 77:341-359. [PMID: 39665487 DOI: 10.1080/01635581.2024.2437833] [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: 07/05/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
This review analyzed existing literature regarding the relationship between different diets and chemotherapy toxicities, as well as the quality of life (QOL) among patients undergoing treatment. It aims to identify the most advantageous diet for cancer patients. PubMed, CINAHL, and Embase were used to select randomized control trials (RCTs) assessing the relationship between a specific diet and chemotherapy toxicities and/or QOL in patients as of October 2023. Out of 1,419 records, 11 RCTs were included. Analyses were stratified by diet type. Pooled odds ratios and 95% confidence intervals (CI) were obtained from the random-effect model using STATA. We included 7 studies testing fasting variations; 1 testing a ketogenic diet; 1 testing a Mediterranean diet; 1 testing a plant-based, high-protein diet; and 1 testing an anti-inflammatory diet. Four fasting studies were in the meta-analysis. The random-effects meta-analysis showed no significant difference in the incidence of chemotherapy toxicities between fasting and non-fasting patients. There is insufficient evidence to determine which dietary intervention is the most advantageous, however, there is evidence that all the diets examined may complement conventional cancer therapy by helping to reduce chemotherapy toxicities. No intervention can be ruled out. More research is needed in this field.
Collapse
Affiliation(s)
- Jessica Abene
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sherilyn Tyburski
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tanja V E Kral
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ryan Quinn
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jie Deng
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Duraj T, Kalamian M, Zuccoli G, Maroon JC, D'Agostino DP, Scheck AC, Poff A, Winter SF, Hu J, Klement RJ, Hickson A, Lee DC, Cooper I, Kofler B, Schwartz KA, Phillips MCL, Champ CE, Zupec-Kania B, Tan-Shalaby J, Serfaty FM, Omene E, Arismendi-Morillo G, Kiebish M, Cheng R, El-Sakka AM, Pflueger A, Mathews EH, Worden D, Shi H, Cincione RI, Spinosa JP, Slocum AK, Iyikesici MS, Yanagisawa A, Pilkington GJ, Chaffee A, Abdel-Hadi W, Elsamman AK, Klein P, Hagihara K, Clemens Z, Yu GW, Evangeliou AE, Nathan JK, Smith K, Fortin D, Dietrich J, Mukherjee P, Seyfried TN. Clinical research framework proposal for ketogenic metabolic therapy in glioblastoma. BMC Med 2024; 22:578. [PMID: 39639257 PMCID: PMC11622503 DOI: 10.1186/s12916-024-03775-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, with a universally lethal prognosis despite maximal standard therapies. Here, we present a consensus treatment protocol based on the metabolic requirements of GBM cells for the two major fermentable fuels: glucose and glutamine. Glucose is a source of carbon and ATP synthesis for tumor growth through glycolysis, while glutamine provides nitrogen, carbon, and ATP synthesis through glutaminolysis. As no tumor can grow without anabolic substrates or energy, the simultaneous targeting of glycolysis and glutaminolysis is expected to reduce the proliferation of most if not all GBM cells. Ketogenic metabolic therapy (KMT) leverages diet-drug combinations that inhibit glycolysis, glutaminolysis, and growth signaling while shifting energy metabolism to therapeutic ketosis. The glucose-ketone index (GKI) is a standardized biomarker for assessing biological compliance, ideally via real-time monitoring. KMT aims to increase substrate competition and normalize the tumor microenvironment through GKI-adjusted ketogenic diets, calorie restriction, and fasting, while also targeting glycolytic and glutaminolytic flux using specific metabolic inhibitors. Non-fermentable fuels, such as ketone bodies, fatty acids, or lactate, are comparatively less efficient in supporting the long-term bioenergetic and biosynthetic demands of cancer cell proliferation. The proposed strategy may be implemented as a synergistic metabolic priming baseline in GBM as well as other tumors driven by glycolysis and glutaminolysis, regardless of their residual mitochondrial function. Suggested best practices are provided to guide future KMT research in metabolic oncology, offering a shared, evidence-driven framework for observational and interventional studies.
Collapse
Affiliation(s)
- Tomás Duraj
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.
| | | | - Giulio Zuccoli
- Neuroradiology, Private Practice, Philadelphia, PA, 19103, USA
| | - Joseph C Maroon
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Adrienne C Scheck
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Phoenix, AZ, 85004, USA
| | - Angela Poff
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Sebastian F Winter
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Jethro Hu
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, 97422, Schweinfurt, Germany
| | | | - Derek C Lee
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Isabella Cooper
- Ageing Biology and Age-Related Diseases Group, School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstr. 48, 5020, Salzburg, Austria
| | - Kenneth A Schwartz
- Department of Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Matthew C L Phillips
- Department of Neurology, Waikato Hospital, Hamilton, 3204, New Zealand
- Department of Medicine, University of Auckland, Auckland, 1142, New Zealand
| | - Colin E Champ
- Exercise Oncology & Resiliency Center and Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA, 15212, USA
| | | | - Jocelyn Tan-Shalaby
- School of Medicine, University of Pittsburgh, Veteran Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Fabiano M Serfaty
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, 20550-170, Brazil
- Serfaty Clínicas, Rio de Janeiro, RJ, 22440-040, Brazil
| | - Egiroh Omene
- Department of Oncology, Cross Cancer Institute, Edmonton, AB, T6G 1Z2, Canada
| | - Gabriel Arismendi-Morillo
- Department of Medicine, Faculty of Health Sciences, University of Deusto, 48007, Bilbao (Bizkaia), Spain
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, 4005, Venezuela
| | | | - Richard Cheng
- Cheng Integrative Health Center, Columbia, SC, 29212, USA
| | - Ahmed M El-Sakka
- Metabolic Terrain Institute of Health, East Congress Street, Tucson, AZ, 85701, USA
| | - Axel Pflueger
- Pflueger Medical Nephrologyand , Internal Medicine Services P.L.L.C, 6 Nelson Road, Monsey, NY, 10952, USA
| | - Edward H Mathews
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0002, South Africa
| | | | - Hanping Shi
- Department of Gastrointestinal Surgery and Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Raffaele Ivan Cincione
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Puglia, Italy
| | - Jean Pierre Spinosa
- Integrative Oncology, Breast and Gynecologic Oncology Surgery, Private Practice, Rue Des Terreaux 2, 1002, Lausanne, Switzerland
| | | | - Mehmet Salih Iyikesici
- Department of Medical Oncology, Altınbaş University Bahçelievler Medical Park Hospital, Istanbul, 34180, Turkey
| | - Atsuo Yanagisawa
- The Japanese College of Intravenous Therapy, Tokyo, 150-0013, Japan
| | | | - Anthony Chaffee
- Department of Neurosurgery, Sir Charles Gairdner Hospital, Perth, 6009, Australia
| | - Wafaa Abdel-Hadi
- Clinical Oncology Department, Cairo University, Giza, 12613, Egypt
| | - Amr K Elsamman
- Neurosurgery Department, Cairo University, Giza, 12613, Egypt
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, 6410 Rockledge Drive, Suite 610, Bethesda, MD, 20817, USA
| | - Keisuke Hagihara
- Department of Advanced Hybrid Medicine, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Zsófia Clemens
- International Center for Medical Nutritional Intervention, Budapest, 1137, Hungary
| | - George W Yu
- George W, Yu Foundation For Nutrition & Health and Aegis Medical & Research Associates, Annapolis, MD, 21401, USA
| | - Athanasios E Evangeliou
- Department of Pediatrics, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, Efkarpia, 56403, Thessaloniki, Greece
| | - Janak K Nathan
- Dr. DY Patil Medical College, Hospital and Research Centre, Pune, Maharashtra, 411018, India
| | - Kris Smith
- Barrow Neurological Institute, Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - David Fortin
- Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
| | - Jorg Dietrich
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | | | | |
Collapse
|
28
|
Bahrami A, Haghighi S, Moghani MM, Khodakarim N, Hejazi E. Fasting mimicking diet during neo-adjuvant chemotherapy in breast cancer patients: a randomized controlled trial study. Front Nutr 2024; 11:1483707. [PMID: 39703333 PMCID: PMC11656309 DOI: 10.3389/fnut.2024.1483707] [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: 08/20/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
Objective Preclinical evidences suggests that while fasting can reduce the side effects and toxicity of chemotherapy, it can make cancer cells more susceptible to chemotherapy. This study aimed to examine the effects of fasting mimicking diet (FMD) during neo-adjuvant chemotherapy in breast cancer (BC) patients. Methods Forty-four newly diagnosed human epidermal growth factor receptor 2-negative (HER2-negative) patients with BC were randomized equally into two groups (22 each), to receive either a fasting mimicking diet (FMD) or their regular diet for 3 days prior to and during neoadjuvant chemotherapy. This FMD was repeated every 3 weeks for 8 cycles. Efficacy, toxicity, hematologic, metabolic, and inflammatory parameters were measured and compared. Results The occurrence of grade III vomiting and neutropenia in the control group was significantly higher than the FMD group (P = <0.001 and p = 0.04 respectively). Erythrocytes (p = 0.01) and neutrophils (p = 0.002) counts were significantly higher in FMD group compared to control group after cycle 8. There was a significant increase in median glucose and median insulin levels (p = 0.01 and p = 0.005, respectively) in the control group between baseline and after cycle 8. While, the median Insulin-like growth factor-1 (IGF1) (p = 0.006) and hs-CRP (p = 0.02) levels were significantly decreased in the FMD group. At the end of study (after cycle 8), the median glucose level was significantly higher in control group (p = 0.008), while the median hs-CRP level was significantly lower in FMD group (p = 0.01). The Miller and Payne pathological response 4/5 (90-100% tumor cell loss) and the radiologically complete or partial response, as measured by MRI or ultrasound before surgery occurred more frequently in FMD group compared to the controls (p = 0.01). Conclusion Fasting mimicking diet was well tolerated during chemotherapy and reduced toxicity of chemotherapy and also, had beneficial effects of some metabolic parameters. Clinical Trial Registration https://irct.behdasht.gov.ir/user/trial/61386/view.
Collapse
Affiliation(s)
- Alireza Bahrami
- Department of Clinical Nutrition and Dietetics, School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Haghighi
- Department of Oncology, Gastroenterology and Liver Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Malekzadeh Moghani
- Department of Radiation Oncology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nastaran Khodakarim
- Department of Hematology & Oncology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Hejazi
- Department of Clinical Nutrition and Dietetics, School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Locasale JW, Goncalves MD, Di Tano M, Burgos-Barragan G. Diet and Cancer Metabolism. Cold Spring Harb Perspect Med 2024; 14:a041549. [PMID: 38621831 PMCID: PMC11610756 DOI: 10.1101/cshperspect.a041549] [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: 04/17/2024]
Abstract
Diet and exercise are modifiable lifestyle factors known to have a major influence on metabolism. Clinical practice addresses diseases of altered metabolism such as diabetes or hypertension by altering these factors. Despite enormous public interest, there are limited defined diet and exercise regimens for cancer patients. Nevertheless, the molecular basis of cancer has converged over the past 15 years on an essential role for altered metabolism in cancer. However, our understanding of the molecular mechanisms that underlie the impact of diet and exercise on cancer metabolism is in its very early stages. In this work, we propose conceptual frameworks for understanding the consequences of diet and exercise on cancer cell metabolism and tumor biology and also highlight recent developments. By advancing our mechanistic understanding, we also discuss actionable ways that such interventions could eventually reach the mainstay of both medical oncology and cancer control and prevention.
Collapse
Affiliation(s)
- Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, Durham, Norh Carolina 27710, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Maira Di Tano
- Division of Endocrinology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Guillermo Burgos-Barragan
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10056, USA
| |
Collapse
|
30
|
Meng Y, Sun J, Zhang G, Yu T, Piao H. Fasting: A Complex, Double-Edged Blade in the Battle Against Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024; 24:1395-1409. [PMID: 39354217 DOI: 10.1007/s12012-024-09925-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
In recent years, there has been a surge in the popularity of fasting as a method to enhance one's health and overall well-being. Fasting is a customary practice characterized by voluntary refraining from consuming food and beverages for a specified duration, ranging from a few hours to several days. The potential advantages of fasting, including enhanced insulin sensitivity, decreased inflammation, and better cellular repair mechanisms, have been well documented. However, the effects of fasting on cancer therapy have been the focus of recent scholarly investigations. Doxorubicin (Dox) is one of the most widely used chemotherapy medications for cancer treatment. Unfortunately, cardiotoxicity, which may lead to heart failure and other cardiovascular issues, has been linked to Dox usage. This study aims to comprehensively examine the possible advantages and disadvantages of fasting concerning Dox-induced cardiotoxicity. Researchers have investigated the potential benefits of fasting in lowering the risk of Dox-induced cardiac damage to solve this problem. Nevertheless, new studies indicate that prolonged alternate-day fasting may adversely affect the heart's capacity to manage the cardiotoxic properties of Dox. Though fasting may benefit overall health, it is essential to proceed cautiously and consider the potential risks in certain circumstances.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Tao Yu
- Department of Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Province Cancer Hospital, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Liaoning Province Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
31
|
Li H, Li C, Ren M, Zhang F, Cao L, Ren K, Ren H. Fasting-Mimicking Diet Prevents Pancreatic Carcinogenesis via Gut Microbiota and Metabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25638-25647. [PMID: 39514436 DOI: 10.1021/acs.jafc.4c06475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The incidence of pancreatic cancer has been increasing globally in recent years and dietary is a well-defined factor contributing to its carcinogenesis. In this study, we showed that in a cerulein-induced KC (Pdx1-cre; LSL-Kras G12D/+) mouse model, a fasting-mimicking diet (FMD)─comprising fasting for 3 days followed by 4 days of refeeding, repeated over three 1-week cycles─significantly retards the progression of pancreatic carcinogenesis. FMD treatment altered gut microbiota, notably boosting butyrate-producing bacteria and elevating butyric acid levels in pancreatic tissues. Furthermore, lysine pan-crotonylation (pan-Kcr) expression was markedly upregulated in pancreatic intraepithelial neoplasia (PanIN) tissues from FMD-treated mice. Treatment of normal pancreatic duct and pancreatic cancer cells with sodium butyrate also upregulated pan-Kcr expression while reducing cell proliferation. Our findings reveal the pivotal role of dietary factors in the carcinogenesis of the pancreas and support further clinical studies of FMD as an antineoplastic therapeutic measure.
Collapse
Affiliation(s)
- Huaxin Li
- Gastrointestinal Cancer Institute/Pancreatic Disease Institute, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chao Li
- Department of Orthopedics, The Affiliated Hospital of Qingdao Binhai University, Qingdao 266404, China
| | - Minghan Ren
- Gastrointestinal Cancer Institute/Pancreatic Disease Institute, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Fang Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Lianjing Cao
- Gastrointestinal Cancer Institute/Pancreatic Disease Institute, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Keyu Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - He Ren
- Gastrointestinal Cancer Institute/Pancreatic Disease Institute, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
32
|
Michenthaler H, Duszka K, Reinisch I, Galhuber M, Moyschewitz E, Stryeck S, Madl T, Prokesch A, Krstic J. Systemic and transcriptional response to intermittent fasting and fasting-mimicking diet in mice. BMC Biol 2024; 22:268. [PMID: 39567986 PMCID: PMC11580389 DOI: 10.1186/s12915-024-02061-2] [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: 02/02/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Dietary restriction (DR) has multiple beneficial effects on health and longevity and can also improve the efficacy of certain therapies. Diets used to instigate DR are diverse and the corresponding response is not uniformly measured. We compared the systemic and liver-specific transcriptional response to intermittent fasting (IF) and commercially available fasting-mimicking diet (FMD) after short- and long-term use in C57BL/6 J mice. RESULTS We show that neither DR regimen causes observable adverse effects in mice. The weight loss was limited to 20% and was quickly compensated during refeeding days. The slightly higher weight loss upon FMD versus IF correlated with stronger fasting response assessed by lower glucose levels and higher ketone body, free fatty acids and especially FGF21 concentrations in blood. RNA sequencing demonstrated similar transcriptional programs in the liver after both regimens, with PPARα signalling as top enriched pathway, while on individual gene level FMD more potently increased gluconeogenesis-related, and PPARα and p53 target gene expression compared to IF. Repeated IF induced similar transcriptional responses as acute IF. However, repeated cycles of FMD resulted in blunted expression of genes involved in ketogenesis and fatty acid oxidation. CONCLUSIONS Short-term FMD causes more pronounced changes in blood parameters and slightly higher weight loss than IF, while both activate similar pathways (particularly PPARα signalling) in the liver. On individual gene level FMD induces a stronger transcriptional response, whereas cyclic application blunts transcriptional upregulation of fatty acid oxidation and ketogenesis only in FMD. Hence, our comparative characterization of IF and FMD protocols renders both as effective DR regimens and serves as resource in the fasting research field.
Collapse
Affiliation(s)
- Helene Michenthaler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
| | - Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Isabel Reinisch
- Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Markus Galhuber
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria
| | - Elisabeth Moyschewitz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
| | - Sarah Stryeck
- Research Centre Pharmaceutical Engineering, Graz University of Technology, Graz, Austria
| | - Tobias Madl
- Division of Medicinal Chemistry, Otto Loewi Research Centre, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Andreas Prokesch
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
33
|
Tang Y, Chen Z, Zuo Q, Kang Y. Regulation of CD8+ T cells by lipid metabolism in cancer progression. Cell Mol Immunol 2024; 21:1215-1230. [PMID: 39402302 PMCID: PMC11527989 DOI: 10.1038/s41423-024-01224-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/22/2024] [Indexed: 11/02/2024] Open
Abstract
Dysregulation of lipid metabolism is a key characteristic of the tumor microenvironment, where tumor cells utilize lipids for proliferation, survival, metastasis, and evasion of immune surveillance. Lipid metabolism has become a critical regulator of CD8+ T-cell-mediated antitumor immunity, with excess lipids in the tumor microenvironment impeding CD8+ T-cell activities. Considering the limited efficacy of immunotherapy in many solid tumors, targeting lipid metabolism to enhance CD8+ T-cell effector functions could significantly improve immunotherapy outcomes. In this review, we examine recent findings on how lipid metabolic processes, including lipid uptake, synthesis, and oxidation, regulate CD8+ T cells within tumors. We also assessed the impact of different lipids on CD8+ T-cell-mediated antitumor immunity, with a particular focus on how lipid metabolism affects mitochondrial function in tumor-infiltrating CD8+ T cells. Furthermore, as cancer is a systemic disease, we examined systemic factors linking lipid metabolism to CD8+ T-cell effector function. Finally, we summarize current therapeutic approaches that target lipid metabolism to increase antitumor immunity and enhance immunotherapy. Understanding the molecular and functional interplay between lipid metabolism and CD8+ T cells offers promising therapeutic opportunities for cancer treatment.
Collapse
Affiliation(s)
- Yong Tang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Ziqing Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
34
|
Dutta A, Chakraborty A, Ghosh T, Kumar A. 5-Fluorouracil induces apoptosis in nutritional deprived hepatocellular carcinoma through mitochondrial damage. Sci Rep 2024; 14:23387. [PMID: 39379402 PMCID: PMC11461840 DOI: 10.1038/s41598-024-73143-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
5-Fluorouracil (5-FU) is the leading chemotherapeutic drug used to treat hepatocellular carcinoma, one of the major cancer diseases after atherosclerosis. Because of chemo-resistance, the success rate of treatment declines with time due to continuous drug exposure. Though autophagy induction is majorly responsible for acquired resistance, the exact role of this evolutionary conserved mechanism is unknown in cancer cell survival and suppression. The usual practice involves the combinatorial use of chemotherapeutic drugs with autophagy inhibitors like Chloroquine and Bafilomycin A, while neglecting the side effects caused by autophagy impairment in healthy cells. Starvation is a well-known physiological inducer of autophagy. In this study, by caloric modulation, we tried to circumvent the resistance imposed by prolonged drug exposure and investigated the effect of 5-FU in nutrient-sufficient and deficient conditions. Our findings show a substantial correlation between autophagy and increased cancer cell death in the presence of 5-FU, with negligible effects on normal cells. Experimental data revealed that nutritional deprivation augmented cell death in the presence of 5-FU through mitochondrial membrane damage and excessive reactive oxygen species (ROS) production, initiating apoptosis. Lipidation study also unveiled that under such combinatorial treatment cellular metabolism shifts from glucose to lipid biosynthesis. Overall, our experimental findings suggest that nutritional deprivation in combination with chemotherapeutic medication can be a new effective strategy to control hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ankita Dutta
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India
| | - Anuja Chakraborty
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India
| | - Tulika Ghosh
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India
| | - Anoop Kumar
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India.
| |
Collapse
|
35
|
Oudmaijer CAJ, Komninos DSJ, Hoeijmakers JHJ, IJzermans JNM, Vermeij WP. Clinical implications of nutritional interventions reducing calories, a systematic scoping review. Clin Nutr ESPEN 2024; 63:427-439. [PMID: 38986906 DOI: 10.1016/j.clnesp.2024.06.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND & AIMS Caloric restriction (CR) constitutes a dietary approach of (temporarily) reducing calorie intake thereby inducing resilience and resistance mechanisms and promoting health. While CR's feasibility and safety have been proven in human trials, its full benefits and translation to different study populations warrants further exploration. METHODS We here conducted a systematic scoping review adhering to Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. RESULTS Our search resulted in 3745 individual records, of which 40 were included. We showed that all studies consistently demonstrated the feasibility and safety of CR-like interventions. The specific effects of nutritional preconditioning vary, further underscoring the need for carefully crafted strategies, according to the intended effect, patient population, and logistical limitations. CONCLUSIONS CR-like interventions (long-term CR or short-term fasting) are feasible in a broad range of patient populations. Whether it has clinical benefit, f.i. reducing treatment-induced side effects and enhancing therapy efficacy, has to be investigated further.
Collapse
Affiliation(s)
- C A J Oudmaijer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Erasmus MC Transplant Institute, Division of Hepatobiliary and Transplantation Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| | - D S J Komninos
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| | - J H J Hoeijmakers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Oncode Institute, Utrecht, The Netherlands; Erasmus MC Cancer Institute, Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, The Netherlands; Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - J N M IJzermans
- Erasmus MC Transplant Institute, Division of Hepatobiliary and Transplantation Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - W P Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Dikeocha IJ, Wardill HR, Coller JK, Bowen JM. Dietary interventions and tumor response to chemotherapy in breast cancer: A comprehensive review of preclinical and clinical data. Clin Nutr ESPEN 2024; 63:462-475. [PMID: 39018241 DOI: 10.1016/j.clnesp.2024.06.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND & AIMS Optimizing treatment efficacy is still a critical part in advancing the treatment of breast cancer. Dietary interventions have drawn significant attention for their potential to increase tumor sensitivity, with a plethora of strategies evaluated both preclinically and clinically. The aim of this paper is to explore these strategies, ranging from entire dietary programs to specific supplements, for their potential to directly enhance tumor sensitivity and chemotherapy adherence. METHODS PubMed, Scopus, Embase and Web of Science databases were searched up to September 2023. In this comprehensive review, preclinical and clinical research on dietary interventions used in conjunction with chemotherapy for breast cancer was examined and synthesized, to identify potential causal mechanisms. RESULTS 42 studies in total were identified and synthesized, 32 pre-clinical and 8 clinical studies. CONCLUSION Although a topic of intense interest, the heterogeneity in approaches has resulted in a large but minimally impactful evidence base, further complicated by a limited understanding of the mechanisms at play. This review highlights the areas for further research to increase opportunities for nutritional-based interventions as adjuvant to chemotherapy for breast cancer.
Collapse
Affiliation(s)
- Ifeoma J Dikeocha
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia.
| | - Hannah R Wardill
- Supportive Oncology Research Group, Precision Cancer Medicine, The South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Janet K Coller
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia
| | - Joanne M Bowen
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
37
|
Kotsifaki A, Maroulaki S, Karalexis E, Stathaki M, Armakolas A. Decoding the Role of Insulin-like Growth Factor 1 and Its Isoforms in Breast Cancer. Int J Mol Sci 2024; 25:9302. [PMID: 39273251 PMCID: PMC11394947 DOI: 10.3390/ijms25179302] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/25/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Insulin-like Growth Factor-1 (IGF-1) is a crucial mitogenic factor with important functions in the mammary gland, mainly through its interaction with the IGF-1 receptor (IGF-1R). This interaction activates a complex signaling network that promotes cell proliferation, epithelial to mesenchymal transition (EMT) and inhibits apoptosis. Despite extensive research, the precise molecular pathways and intracellular mechanisms activated by IGF-1, in cancer, remain poorly understood. Recent evidence highlights the essential roles of IGF-1 and its isoforms in breast cancer (BC) development, progression, and metastasis. The peptides that define the IGF-1 isoforms-IGF-1Ea, IGF-1Eb, and IGF-1Ec-act as key points of convergence for various signaling pathways that influence the growth, metastasis and survival of BC cells. The aim of this review is to provide a detailed exami-nation of the role of the mature IGF-1 and its isoforms in BC biology and their potential use as possible therapeutical targets.
Collapse
Affiliation(s)
- Amalia Kotsifaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sousanna Maroulaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efthymios Karalexis
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Martha Stathaki
- Surgical Clinic, "Elena Venizelou" General Hospital, 11521 Athens, Greece
| | - Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
38
|
Vernieri C, Ligorio F, Tripathy D, Longo VD. Cyclic fasting-mimicking diet in cancer treatment: Preclinical and clinical evidence. Cell Metab 2024; 36:1644-1667. [PMID: 39059383 DOI: 10.1016/j.cmet.2024.06.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
In preclinical tumor models, cyclic fasting and fasting-mimicking diets (FMDs) produce antitumor effects that become synergistic when combined with a wide range of standard anticancer treatments while protecting normal tissues from treatment-induced adverse events. More recently, results of phase 1/2 clinical trials showed that cyclic FMD is safe, feasible, and associated with positive metabolic and immunomodulatory effects in patients with different tumor types, thus paving the way for larger clinical trials to investigate FMD anticancer activity in different clinical contexts. Here, we review the tumor-cell-autonomous and immune-system-mediated mechanisms of fasting/FMD antitumor effects, and we critically discuss new metabolic interventions that could synergize with nutrient starvation to boost its anticancer activity and prevent or reverse tumor resistance while minimizing toxicity to patients. Finally, we highlight potential future applications of FMD approaches in combination with standard anticancer strategies as well as strategies to implement the design and conduction of clinical trials.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology and Hematology-Oncology Department, University of Milan, 20122 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, 20139 Milan, Italy.
| | - Francesca Ligorio
- Medical Oncology and Hematology-Oncology Department, University of Milan, 20122 Milan, Italy; Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1354, 1515 Holcombe Blvd, Houston, TX 77030-4009, USA
| | - Valter D Longo
- IFOM ETS, the AIRC Institute of Molecular Oncology, 20139 Milan, Italy; Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
39
|
Koppold DA, Breinlinger C, Hanslian E, Kessler C, Cramer H, Khokhar AR, Peterson CM, Tinsley G, Vernieri C, Bloomer RJ, Boschmann M, Bragazzi NL, Brandhorst S, Gabel K, Goldhamer AC, Grajower MM, Harvie M, Heilbronn L, Horne BD, Karras SN, Langhorst J, Lischka E, Madeo F, Mitchell SJ, Papagiannopoulos-Vatopaidinos IE, Papagiannopoulou M, Pijl H, Ravussin E, Ritzmann-Widderich M, Varady K, Adamidou L, Chihaoui M, de Cabo R, Hassanein M, Lessan N, Longo V, Manoogian ENC, Mattson MP, Muhlestein JB, Panda S, Papadopoulou SK, Rodopaios NE, Stange R, Michalsen A. International consensus on fasting terminology. Cell Metab 2024; 36:1779-1794.e4. [PMID: 39059384 PMCID: PMC11504329 DOI: 10.1016/j.cmet.2024.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/16/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Although fasting is increasingly applied for disease prevention and treatment, consensus on terminology is lacking. Using Delphi methodology, an international, multidisciplinary panel of researchers and clinicians standardized definitions of various fasting approaches in humans. Five online surveys and a live online conference were conducted with 38 experts, 25 of whom completed all 5 surveys. Consensus was achieved for the following terms: "fasting" (voluntary abstinence from some or all foods or foods and beverages), "modified fasting" (restriction of energy intake to max. 25% of energy needs), "fluid-only fasting," "alternate-day fasting," "short-term fasting" (lasting 2-3 days), "prolonged fasting" (≥4 consecutive days), and "religious fasting." "Intermittent fasting" (repetitive fasting periods lasting ≤48 h), "time-restricted eating," and "fasting-mimicking diet" were discussed most. This study provides expert recommendations on fasting terminology for future research and clinical applications, facilitating communication and cross-referencing in the field.
Collapse
Affiliation(s)
- Daniela A Koppold
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Department of Internal Medicine and Nature-Based Therapies, Immanuel Hospital Berlin, 14109 Berlin, Germany; Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.
| | - Carolin Breinlinger
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany.
| | - Etienne Hanslian
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Christian Kessler
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Department of Internal Medicine and Nature-Based Therapies, Immanuel Hospital Berlin, 14109 Berlin, Germany
| | - Holger Cramer
- Institute for General Practice and Interprofessional Care, University Hospital Tübingen, Tübingen, Germany; Robert Bosch Center for Integrative Medicine and Health, Bosch Health Campus, Stuttgart, Germany
| | - Anika Rajput Khokhar
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Courtney M Peterson
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Grant Tinsley
- Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Richard J Bloomer
- College of Health Sciences, The University of Memphis, Memphis, TN 38152, USA
| | - Michael Boschmann
- Experimental & Clinical Research Center - A joint co-operation between Charité Universitätsmedizin und Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Clinical Research Unit, Berlin, Germany
| | - Nicola L Bragazzi
- Department of Mathematics and Statistics, Laboratory for Industrial and Applied Mathematics (LIAM), York University, Toronto, ON, Canada
| | - Sebastian Brandhorst
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kelsey Gabel
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, 1919 West Taylor Street, Chicago, IL 60612, USA
| | - Alan C Goldhamer
- TrueNorth Health Foundation, Santa Rosa, CA 95404, USA; TrueNorth Health Center, Santa Rosa, CA 95404, USA
| | - Martin M Grajower
- Division of Endocrinology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Michelle Harvie
- Prevent Breast Cancer Research Unit, The Nightingale Centre, Manchester University NHS Foundation Trust, Manchester, England; Division of Cancer Sciences, The University of Manchester, Manchester, England
| | - Leonie Heilbronn
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia; Nutrition, Metabolism & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Benjamin D Horne
- Intermountain Medical Center Heart Institute, Salt Lake City, UT, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Spyridon N Karras
- Laboratory of Biological Chemistry, Medical School, Aristotle University, 54636 Thessaloniki, Greece
| | - Jost Langhorst
- Department for Internal and Integrative Medicine, Sozialstiftung Bamberg Hospital, Bamberg, Germany; Department for Integrative Medicine, University of Duisburg-Essen, Medical Faculty, Bamberg, Germany
| | - Eva Lischka
- Klinik Buchinger Wilhelmi, Überlingen, Germany
| | - Frank Madeo
- BioTechMed Graz, Graz, Austria; Institute of Molecular Biosciences, University of Graz, Graz, Austria; Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Sarah J Mitchell
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | | | | | - Hanno Pijl
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Martha Ritzmann-Widderich
- Praxis für Ernährungsmedizin und Prävention in Rottweil, Hochbrücktorstraße 22, 78628 Rottweil, Germany
| | - Krista Varady
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, 1919 West Taylor Street, Chicago, IL 60612, USA
| | - Lilian Adamidou
- Department of Dietetics and Nutrition, AHEPA University Hospital, Thessaloniki, Greece
| | - Melika Chihaoui
- Department of Endocrinology, University Hospital La Rabta, Faculty of medicine of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Mohamed Hassanein
- Department of Endocrinology and Diabetes, Dubai Hospital, Dubai Academic Health Cooperation, United Arab Emirates
| | - Nader Lessan
- The Research Institute, Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates
| | - Valter Longo
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy; Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Emily N C Manoogian
- Regulatory Biology Department, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Satchidananda Panda
- Regulatory Biology Department, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sousana K Papadopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57001 Thessaloniki, Greece
| | - Nikolaos E Rodopaios
- Department of Social Medicine, Preventive Medicine and Nutrition Clinic, School of Medicine, University of Crete, Voutes, 71003 Iraklion, Greece
| | - Rainer Stange
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Department of Internal Medicine and Nature-Based Therapies, Immanuel Hospital Berlin, 14109 Berlin, Germany
| | - Andreas Michalsen
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Department of Internal Medicine and Nature-Based Therapies, Immanuel Hospital Berlin, 14109 Berlin, Germany
| |
Collapse
|
40
|
Caron JP, Ernyey H, Rosenthal MD. Can caloric restriction improve outcomes of elective surgeries? JPEN J Parenter Enteral Nutr 2024; 48:646-657. [PMID: 38802250 DOI: 10.1002/jpen.2642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Energy restriction (ER) is a nutrition method to reduce the amount of energy intake while maintaining adequate nutrition. In clinical medicine, applications of ER have been implicated in longevity, mortality, metabolic, immune, and psychological health. However, there are limited studies showing the clinical benefit of ER within the immediate surgical setting. A specific, clinically oriented summary of the potential applications of ER is needed to optimize surgery outcomes for patients. The purpose of this article is to examine how ER can be used for perioperative optimization to improve outcomes for the patient and surgeon. It will also explore how these outcomes can feasibly fit in with enhanced recovery after surgery protocols and can be used as a method for nutrition optimization in surgery. Despite evidence of caloric restriction improving outcomes in critically ill surgical patients, there is not enough evidence to conclude that ER, perioperatively across noncritically ill cohorts, improves postoperative morbidity and mortality in elective surgeries. Nevertheless, a contemporary account of how ER techniques may have a significant role in reducing risk factors of adverse surgical outcomes in this cohort, for example, by encouraging preoperative weight loss contributing to decreased operating times, is reviewed.
Collapse
Affiliation(s)
| | - Helen Ernyey
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
41
|
Tizazu AM. Fasting and calorie restriction modulate age-associated immunosenescence and inflammaging. Aging Med (Milton) 2024; 7:499-509. [PMID: 39234195 PMCID: PMC11369340 DOI: 10.1002/agm2.12342] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Aging is a multifaceted process impacting cells, tissues, organs, and organ systems of the body. Like other systems, aging affects both the adaptive and the innate components of the immune system, a phenomenon known as immunosenescence. The deregulation of the immune system puts elderly individuals at higher risk of infection, lower response to vaccines, and increased incidence of cancer. In the Western world, overnutrition has increased the incidence of obesity (linked with chronic inflammation) which increases the risk of metabolic syndrome, cardiovascular disease, and cancer. Aging is also associated with inflammaging a sterile chronic inflammation that predisposes individuals to age-associated disease. Genetic manipulation of the nutrient-sensing pathway, fasting, and calorie restriction (CR) has been shown to increase the lifespan of model organisms. As well in humans, fasting and CR have also been shown to improve different health parameters. Yet the direct effect of fasting and CR on the aging immune system needs to be further explored. Identifying the effect of fasting and CR on the immune system and how it modulates different parameters of immunosenescence could be important in designing pharmacological or nutritional interventions that slow or revert immunosenescence and strengthen the immune system of elderly individuals. Furthermore, clinical intervention can also be planned, by incorporating fasting or CR with medication, chemotherapy, and vaccination regimes. This review discusses age-associated changes in the immune system and how these changes are modified by fasting and CR which add information on interventions that promote healthy aging and longevity in the growing aging population.
Collapse
Affiliation(s)
- Anteneh Mehari Tizazu
- Department of Microbiology, Immunology, and Parasitology, School of MedicineSt. Paul's Hospital Millennium Medical CollegeAddis AbabaEthiopia
| |
Collapse
|
42
|
De-Leon-Covarrubias UE, Perez-Trujillo JJ, Villa-Cedillo SA, Martinez-Perez AG, Montes-de-Oca-Saucedo CR, Loera-Arias MDJ, Garcia-Garcia A, Saucedo-Cardenas O, Montes-de-Oca-Luna R. Unlocking the Potential: Caloric Restriction, Caloric Restriction Mimetics, and Their Impact on Cancer Prevention and Treatment. Metabolites 2024; 14:418. [PMID: 39195514 DOI: 10.3390/metabo14080418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024] Open
Abstract
Caloric restriction (CR) and its related alternatives have been shown to be the only interventions capable of extending lifespan and decreasing the risk of cancer, along with a reduction in burden in pre-clinical trials. Nevertheless, the results from clinical trials have not been as conclusive as the pre-clinical results. Recognizing the challenges associated with long-term fasting, the application of caloric restriction mimetics (CRMs), pharmacological agents that mimic the molecular effects of CR, to harness the potential benefits while overcoming the practical limitations of fasting has resulted in an interesting alternative. This review synthesizes the findings of diverse clinical trials evaluating the safety and efficacy of CR and CRMs. In dietary interventions, a fast-mimicking diet was the most tolerated to reduce tumoral growth markers and chemotherapy side effects. CRMs were well tolerated, and metformin and aspirin showed the most promising effect in reducing cancer risk in a selected group of patients. The application of CR and/or CRMs shows promising effects in anti-cancer therapy; however, there is a need for more evidence to safely include these interventions in standard-of-care therapies.
Collapse
Affiliation(s)
| | - Jose Juan Perez-Trujillo
- Histology Department, Faculty of Medicine, Universidad Autonoma de Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Sheila Adela Villa-Cedillo
- Histology Department, Faculty of Medicine, Universidad Autonoma de Nuevo Leon (UANL), Monterrey 64460, Mexico
| | | | | | - Maria de Jesus Loera-Arias
- Histology Department, Faculty of Medicine, Universidad Autonoma de Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Aracely Garcia-Garcia
- Histology Department, Faculty of Medicine, Universidad Autonoma de Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Odila Saucedo-Cardenas
- Histology Department, Faculty of Medicine, Universidad Autonoma de Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Roberto Montes-de-Oca-Luna
- Histology Department, Faculty of Medicine, Universidad Autonoma de Nuevo Leon (UANL), Monterrey 64460, Mexico
| |
Collapse
|
43
|
Pan T, Tang L, Chu R, Zheng S, Wang J, Yang Y, Wang W, He J. Microfluidic-Enabled Assembly of Multicomponent Artificial Organelle for Synergistic Tumor Starvation Therapy. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39069732 DOI: 10.1021/acsami.4c07962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Artificial organelles (AOs) encapsulating enzymes are engineered to facilitate biocatalytic reactions for exerting therapeutic effects in various diseases. Exploiting the confinement effect, these catalytic properties exhibit significant enhancements without being influenced by the surrounding medium, enabling more efficient cascade reactions. In this study, we present a novel approach for synergistic tumor starvation therapy by developing multicomponent artificial organelles that combine enzymatic oncotherapy with chemotherapy. The construction process involves a microfluidic-based approach that enables the encapsulation of cationic cores containing doxorubicin (DOX), electrostatic adsorption of cascade enzymes, and surface assembly of the protective lipid membrane. Additionally, these multicomponent AOs possess multicompartment structures that enable the separation and sequential release of each component. By coencapsulating enzymes and chemotherapeutic agent DOX within AOs, we achieve enhanced enzymatic cascade reactions (ECR) and improved intrinsic permeability of DOX due to spatial confinement. Furthermore, exceptional therapeutic effects on 4T1 xenograft tumors are observed, demonstrating the feasibility of utilizing AOs as biomimetic implants in living organisms. This innovative approach that combines starvation therapy with chemotherapy using multicompartment AOs represents a promising paradigm in the field of precise cancer therapy.
Collapse
Affiliation(s)
- Ting Pan
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, P. R. China
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Runxuan Chu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, P. R. China
| | - Shumin Zheng
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, P. R. China
| | - Junji Wang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, P. R. China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, P. R. China
| |
Collapse
|
44
|
Khalifa A, Guijarro A, Nencioni A. Advances in Diet and Physical Activity in Breast Cancer Prevention and Treatment. Nutrients 2024; 16:2262. [PMID: 39064705 PMCID: PMC11279876 DOI: 10.3390/nu16142262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
There is currently a growing interest in diets and physical activity patterns that may be beneficial in preventing and treating breast cancer (BC). Mounting evidence indicates that indeed, the so-called Mediterranean diet (MedDiet) and regular physical activity likely both help reduce the risk of developing BC. For those who have already received a BC diagnosis, these interventions may decrease the risk of tumor recurrence after treatment and improve quality of life. Studies also show the potential of other dietary interventions, including fasting or modified fasting, calorie restriction, ketogenic diets, and vegan or plant-based diets, to enhance the efficacy of BC therapies. In this review article, we discuss the biological rationale for utilizing these dietary interventions and physical activity in BC prevention and treatment. We highlight published and ongoing clinical studies that have applied these lifestyle interventions to BC patients. This review offers valuable insights into the potential application of these dietary interventions and physical activity as complimentary therapies in BC management.
Collapse
Affiliation(s)
- Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy;
| | - Ana Guijarro
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy;
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
45
|
Gabriel S, Myers TR, Thompson N, Goldhamer AC. Prolonged water-only fasting in the management of low-grade follicular lymphoma: a case series. J Med Case Rep 2024; 18:302. [PMID: 38956708 PMCID: PMC11221076 DOI: 10.1186/s13256-024-04609-w] [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: 11/09/2023] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Follicular lymphoma typically follows an indolent and relapsing course often requiring several treatment cycles to achieve remission. Some patients opt to use complementary and alternative therapies particularly when observation is a treatment option. CASE PRESENTATION Here we present a case series of three patients, a 50-year-old, White, Hispanic female, 56-year-old, White, non-Hispanic male, and 49-year-old, White, non-Hispanic male, who elected to undergo one or more prolonged water-only fasting and refeeding interventions to manage low to intermediate grade follicular lymphoma. Fasting was well tolerated in each patient. Each patient also experienced a reduction in the size and avidity of hypermetabolic lymph nodes as independently determined by their respective oncologists. CONCLUSION The reported cases demonstrate positive outcomes in low-grade follicular lymphoma coinciding with prolonged water-only fasting and exclusively whole-plant-food dietary interventions. These findings highlight the potential of such interventions and warrant further exploration through preliminary observational research.
Collapse
|
46
|
Da Prat V, Pravettoni G, Casirati A, Marzorati C, Pedrazzoli P, Caccialanza R. Anticancer restrictive diets and the risk of psychological distress: Review and perspectives. Cancer Med 2024; 13:e7329. [PMID: 38970205 PMCID: PMC11226408 DOI: 10.1002/cam4.7329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/03/2024] [Accepted: 05/12/2024] [Indexed: 07/08/2024] Open
Abstract
INTRODUCTION The most studied anticancer restrictive diets include fasting, fasting-mimicking diets (FMDs) and ketogenic diets (KDs). Besides the current lack of established clinical benefit and the significant risk of malnutrition and micronutrient deficiencies, dietary restrictions in cancer patients might have relevant psychological effects. MATERIALS AND METHODS We reviewed the randomized and non-randomized controlled clinical trials (CCTs) reporting data on the psychological impact of fasting, FMDs and KDs in cancer patients. We excluded trials on restrictive diets performed for weight reduction in obese or overweight patients, studies on dietary restrictions lasting less than 24 h, and studies on fasting related to cultural or religious beliefs. RESULTS Three CCTs on fasting or FMDs and eight CCTs on KDs in cancer patients were included. In terms of diet-related distress, emotional, social, and family well-being, none of these studies showed a detrimental impact of fasting, FMDs and KDs. However, clinical trials specifically assessing the psychological aspects in the long term are lacking. CONCLUSIONS AND PERSPECTIVES In the absence of a conclusive evidence on the clinical benefits of restrictive diets, which carry significant risks especially if unsupervised, further studies are needed to clarify their psychological impact in cancer patients. Multidisciplinary approaches including psychological evaluations should be used to ameliorate patient selection for clinical trials, identify early distress symptoms, and increase patient compliance to dietary recommendations.
Collapse
Affiliation(s)
- Valentina Da Prat
- Clinical Nutrition and Dietetics UnitFondazione IRCCS Policlinico San MatteoPaviaItaly
| | - Gabriella Pravettoni
- Applied Research Division for Cognitive and Psychological ScienceIstituto Europeo di Oncologia IRCCSMilanItaly
- Department of Oncology and Hemato‐OncologyUniversity of MilanMilanItaly
| | - Amanda Casirati
- Clinical Nutrition and Dietetics UnitFondazione IRCCS Policlinico San MatteoPaviaItaly
| | - Chiara Marzorati
- Applied Research Division for Cognitive and Psychological ScienceIstituto Europeo di Oncologia IRCCSMilanItaly
| | - Paolo Pedrazzoli
- Department of OncologyFondazione IRCCS Policlinico San MatteoPaviaItaly
- Department of Internal Medicine and Medical TherapyUniversity of PaviaPaviaItaly
| | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics UnitFondazione IRCCS Policlinico San MatteoPaviaItaly
| |
Collapse
|
47
|
Son DS, Done KA, Son J, Izban MG, Virgous C, Lee ES, Adunyah SE. Intermittent Fasting Attenuates Obesity-Induced Triple-Negative Breast Cancer Progression by Disrupting Cell Cycle, Epithelial-Mesenchymal Transition, Immune Contexture, and Proinflammatory Signature. Nutrients 2024; 16:2101. [PMID: 38999849 PMCID: PMC11243652 DOI: 10.3390/nu16132101] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Obesity is associated with one-fifth of cancer deaths, and breast cancer is one of the obesity-related cancers. Triple-negative breast cancer (TNBC) lacks estrogen and progesterone receptors and human epidermal growth factor receptor 2, leading to the absence of these therapeutic targets, followed by poor overall survival. We investigated if obesity could hasten TNBC progression and intermittent fasting (IF) could attenuate the progression of obesity-related TNBC. Our meta-analysis of the TNBC outcomes literature showed that obesity led to poorer overall survival in TNBC patients. Fasting-mimicking media reduced cell proliferation disrupted the cell cycle, and decreased cell migration and invasion. IF decreased body weight in obese mice but no change in normal mice. Obese mice exhibited elevated plasma glucose and cholesterol levels, increased tumor volume and weight, and enhanced macrophage accumulation in tumors. The obesity-exacerbated TNBC progression was attenuated after IF, which decreased cyclin B1 and vimentin levels and reduced the proinflammatory signature in the obesity-associated tumor microenvironment. IF attenuated obesity-induced TNBC progression through reduced obesity and tumor burdens in cell and animal experiments, supporting the potential of a cost-effective adjuvant IF therapy for TNBC through lifestyle change. Further evidence is needed of these IF benefits in TNBC, including from human clinical trials.
Collapse
Affiliation(s)
- Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Kaitlyn A. Done
- Biochemistry Program, College of Arts and Sciences, Spelman College, Atlanta, GA 30314, USA
| | - Jubin Son
- Neuroscience Program, College of Arts and Sciences, The University of Tennessee, Knoxville, TN 37996, USA
| | - Michael G. Izban
- Pathology, Anatomy and Cell Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Carlos Virgous
- Animal Core Facility, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA;
| | - Samuel E. Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
48
|
Luo M, Wang Q, Sun Y, Jiang Y, Wang Q, Gu Y, Hu Z, Chen Q, Xu J, Chen S, Hou T, Feng L. Fasting-mimicking diet remodels gut microbiota and suppresses colorectal cancer progression. NPJ Biofilms Microbiomes 2024; 10:53. [PMID: 38918380 PMCID: PMC11199600 DOI: 10.1038/s41522-024-00520-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
The progression of colorectal cancer is closely associated with diet. Fasting-mimicking diet (FMD) is a promising type of dietary intervention that have beneficial effects in the prevention and treatment of various cancers. We investigated the therapeutic effect of 4-day FMD against colorectal cancer in mice through immune cell analysis, microbiota composition analysis and anti-PD-1 treatment. These FMD cycles effectively suppressed colorectal cancer growth, reduced cell proliferation and angiogenesis, increased tumor-infiltration lymphocytes especially CD8+T cells. FMD stimulated protective gut microbiota, especially Lactobacillus. Supplementation of Lactobacillus johnsonii induced similar results as FMD intervention, which also suppressed tumor growth and increased CD45+ and CD8+ T cells. Additionally, FMD synthesizing with anti-PD-1 therapy effectively inhibited CRC progression. These findings suggest that Lactobacillus. johnsonii is necessary for the anticancer process of FMD in CRC. FMD through its effects on both gut microbiota and immune system, effectively suppressed colorectal cancer progression in mouse model.
Collapse
Affiliation(s)
- Man Luo
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Qingyi Wang
- Medical School of Zhejiang University, Hangzhou, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yong Sun
- Medical School of Zhejiang University, Hangzhou, China
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Jiang
- Medical School of Zhejiang University, Hangzhou, China
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qiwen Wang
- Medical School of Zhejiang University, Hangzhou, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yanrou Gu
- Wenzhou Medical University, Wenzhou, China
| | - Zhefang Hu
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Qianyi Chen
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Jilei Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Tongyao Hou
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.
| | - Lijun Feng
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.
| |
Collapse
|
49
|
Boufaied N, Chetta P, Hallal T, Cacciatore S, Lalli D, Luthold C, Homsy K, Imada EL, Syamala S, Photopoulos C, Di Matteo A, de Polo A, Storaci AM, Huang Y, Giunchi F, Sheridan PA, Michelotti G, Nguyen QD, Zhao X, Liu Y, Davicioni E, Spratt DE, Sabbioneda S, Maga G, Mucci LA, Ghigna C, Marchionni L, Butler LM, Ellis L, Bordeleau F, Loda M, Vaira V, Labbé DP, Zadra G. Obesogenic High-Fat Diet and MYC Cooperate to Promote Lactate Accumulation and Tumor Microenvironment Remodeling in Prostate Cancer. Cancer Res 2024; 84:1834-1855. [PMID: 38831751 PMCID: PMC11148549 DOI: 10.1158/0008-5472.can-23-0519] [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: 02/17/2023] [Revised: 12/29/2023] [Accepted: 04/05/2024] [Indexed: 06/05/2024]
Abstract
Cancer cells exhibit metabolic plasticity to meet oncogene-driven dependencies while coping with nutrient availability. A better understanding of how systemic metabolism impacts the accumulation of metabolites that reprogram the tumor microenvironment (TME) and drive cancer could facilitate development of precision nutrition approaches. Using the Hi-MYC prostate cancer mouse model, we demonstrated that an obesogenic high-fat diet (HFD) rich in saturated fats accelerates the development of c-MYC-driven invasive prostate cancer through metabolic rewiring. Although c-MYC modulated key metabolic pathways, interaction with an obesogenic HFD was necessary to induce glycolysis and lactate accumulation in tumors. These metabolic changes were associated with augmented infiltration of CD206+ and PD-L1+ tumor-associated macrophages (TAM) and FOXP3+ regulatory T cells, as well as with the activation of transcriptional programs linked to disease progression and therapy resistance. Lactate itself also stimulated neoangiogenesis and prostate cancer cell migration, which were significantly reduced following treatment with the lactate dehydrogenase inhibitor FX11. In patients with prostate cancer, high saturated fat intake and increased body mass index were associated with tumor glycolytic features that promote the infiltration of M2-like TAMs. Finally, upregulation of lactate dehydrogenase, indicative of a lactagenic phenotype, was associated with a shorter time to biochemical recurrence in independent clinical cohorts. This work identifies cooperation between genetic drivers and systemic metabolism to hijack the TME and promote prostate cancer progression through oncometabolite accumulation. This sets the stage for the assessment of lactate as a prognostic biomarker and supports strategies of dietary intervention and direct lactagenesis blockade in treating advanced prostate cancer. SIGNIFICANCE Lactate accumulation driven by high-fat diet and MYC reprograms the tumor microenvironment and promotes prostate cancer progression, supporting the potential of lactate as a biomarker and therapeutic target in prostate cancer. See related commentary by Frigo, p. 1742.
Collapse
Affiliation(s)
- Nadia Boufaied
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Paolo Chetta
- University of Milan, Residency Program in Pathology, Milan, Italy
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Stefano Cacciatore
- Bionformatics Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | - Daniela Lalli
- Department of Science and Technological Innovation, University of Piemonte Orientale “A. Avogadro,” Alessandria, Italy
| | - Carole Luthold
- CHU de Québec-Université Laval Research Center (Oncology Division) and Cancer Research Center, Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Québec, Canada
| | - Kevin Homsy
- CHU de Québec-Université Laval Research Center (Oncology Division) and Cancer Research Center, Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Québec, Canada
| | - Eddie L. Imada
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York Presbyterian-Weill Cornell Campus, New York, New York
| | - Sudeepa Syamala
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Cornelia Photopoulos
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Anna Di Matteo
- Institute of Molecular Genetics, National Research Council (CNR-IGM), Pavia, Italy
| | - Anna de Polo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | | | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | | | - Quang-De Nguyen
- Department of Imaging, Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Xin Zhao
- Veracyte, South San Francisco, California
| | - Yang Liu
- Veracyte, South San Francisco, California
| | | | - Daniel E. Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Simone Sabbioneda
- Institute of Molecular Genetics, National Research Council (CNR-IGM), Pavia, Italy
| | - Giovanni Maga
- Institute of Molecular Genetics, National Research Council (CNR-IGM), Pavia, Italy
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Claudia Ghigna
- Institute of Molecular Genetics, National Research Council (CNR-IGM), Pavia, Italy
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York Presbyterian-Weill Cornell Campus, New York, New York
| | - Lisa M. Butler
- South Australian Immunogenomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Leigh Ellis
- Department of Surgery, Center for Prostate Disease Research, Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland
| | - François Bordeleau
- CHU de Québec-Université Laval Research Center (Oncology Division) and Cancer Research Center, Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Québec, Canada
- Department of Molecular Biology, Clinical Biochemistry, and Pathology, Laval University, Québec, Canada
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York Presbyterian-Weill Cornell Campus, New York, New York
| | - Valentina Vaira
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
- Division of Urology, Department of Surgery, McGill University, Montréal, Québec, Canada
| | - Giorgia Zadra
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Institute of Molecular Genetics, National Research Council (CNR-IGM), Pavia, Italy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Li Sucholeiki R, Propst CL, Hong DS, George GC. Intermittent fasting and its impact on toxicities, symptoms and quality of life in patients on active cancer treatment. Cancer Treat Rev 2024; 126:102725. [PMID: 38574507 PMCID: PMC11614448 DOI: 10.1016/j.ctrv.2024.102725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Intermittent fasting is a dietary intervention that is increasingly being tested for positive outcomes in patients receiving cancer treatment. In this review, we examine the impact of intermittent fasting on symptoms, toxicities, and quality of life in patients undergoing cancer therapy and highlight unmet investigative areas to prompt future research. While current evidence is preliminary and conclusions mixed, some promising clinical studies suggest that intermittent fasting interventions may improve fatigue and reduce gastrointestinal toxicities in certain patients with cancer. Emerging clinical evidence also demonstrates that intermittent fasting may reduce off-target DNA damage, and induce favorable cellular-level immune remodeling. Furthermore, intermittent fasting has the potential to lower hyperglycemia and the ratio of fat to lean body mass, which may benefit patients at risk of hyperglycemia and weight-related adverse effects of some common pharmacological cancer treatments. Larger controlled studies are necessary to evaluate intermittent fasting in relation to these endpoints and determine the effectiveness of intermittent fasting as an adjunct intervention during cancer care. Future cancer trials should evaluate intermittent fasting diets in the context of multimodal diet, exercise, and nutrition strategies, and also evaluate the impact of intermittent fasting on other important areas such as the circadian system and the gut microbiome.
Collapse
Affiliation(s)
- Robert Li Sucholeiki
- University of Chicago, M. D. Anderson Cancer Center, United States; The University of Texas M. D. Anderson Cancer Center, United States
| | - Casey L Propst
- University of Chicago, M. D. Anderson Cancer Center, United States; The University of Texas M. D. Anderson Cancer Center, United States
| | - David S Hong
- The University of Texas M. D. Anderson Cancer Center, United States
| | - Goldy C George
- The University of Texas M. D. Anderson Cancer Center, United States.
| |
Collapse
|