51
|
Lai C, Xu L, Dai S. The nuclear export protein exportin-1 in solid malignant tumours: From biology to clinical trials. Clin Transl Med 2024; 14:e1684. [PMID: 38783482 PMCID: PMC11116501 DOI: 10.1002/ctm2.1684] [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: 12/07/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Exportin-1 (XPO1), a crucial protein regulating nuclear-cytoplasmic transport, is frequently overexpressed in various cancers, driving tumor progression and drug resistance. This makes XPO1 an attractive therapeutic target. Over the past few decades, the number of available nuclear export-selective inhibitors has been increasing. Only KPT-330 (selinexor) has been successfully used for treating haematological malignancies, and KPT-8602 (eltanexor) has been used for treating haematologic tumours in clinical trials. However, the use of nuclear export-selective inhibitors for the inhibition of XPO1 expression has yet to be thoroughly investigated in clinical studies and therapeutic outcomes for solid tumours. METHODS We collected numerous literatures to explain the efficacy of XPO1 Inhibitors in preclinical and clinical studies of a wide range of solid tumours. RESULTS In this review, we focus on the nuclear export function of XPO1 and results from clinical trials of its inhibitors in solid malignant tumours. We summarized the mechanism of action and therapeutic potential of XPO1 inhibitors, as well as adverse effects and response biomarkers. CONCLUSION XPO1 inhibition has emerged as a promising therapeutic strategy in the fight against cancer, offering a novel approach to targeting tumorigenic processes and overcoming drug resistance. SINE compounds have demonstrated efficacy in a wide range of solid tumours, and ongoing research is focused on optimizing their use, identifying response biomarkers, and developing effective combination therapies. KEY POINTS Exportin-1 (XPO1) plays a critical role in mediating nucleocytoplasmic transport and cell cycle. XPO1 dysfunction promotes tumourigenesis and drug resistance within solid tumours. The therapeutic potential and ongoing researches on XPO1 inhibitors in the treatment of solid tumours. Additional researches are essential to address safety concerns and identify biomarkers for predicting patient response to XPO1 inhibitors.
Collapse
Affiliation(s)
- Chuanxi Lai
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| | - Lingna Xu
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| | - Sheng Dai
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
52
|
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
|
53
|
Wang Q, Xu J, Luo M, Jiang Y, Gu Y, Wang Q, He J, Sun Y, Lin Y, Feng L, Chen S, Hou T. Fasting mimicking diet extends lifespan and improves intestinal and cognitive health. Food Funct 2024; 15:4503-4514. [PMID: 38567489 DOI: 10.1039/d4fo00483c] [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: 04/04/2024]
Abstract
Caloric restriction is an effective means of extending a healthy lifespan. Fasting mimicking diet (FMD) is a growing pattern of caloric restriction. We found that FMD significantly prolonged the lifespan of prematurely aging mice. In naturally aging mice, FMD improved cognitive and intestinal health. Through a series of behavioral experiments, we found that FMD relieved anxiety and enhanced cognition in aged mice. In the intestine, the FMD cycles enhanced the barrier function, reduced senescence markers, and maintained T cell naïve-memory balance in the lamina propria mucosa. To further explore the causes of immune alterations, we examined changes in the stool microbiota using 16S rRNA sequencing. We found that FMD remodeled gut bacterial composition and significantly expanded the abundance of Lactobacillus johnsonii. Our research revealed that FMD has in-depth investigative value as an anti-aging intervention for extending longevity and improving cognition, intestinal function, and gut microbiota composition.
Collapse
Affiliation(s)
- Qingyi Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Jilei Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Man Luo
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yao Jiang
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
- Department of Gastroenterology, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Yanrou Gu
- Department of Gastroenterology, Wenzhou No. 3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Qiwen Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Jiamin He
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Yong Sun
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
- Department of Gastroenterology, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Yifeng Lin
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
- Department of Gastroenterology, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Lijun Feng
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Tongyao Hou
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| |
Collapse
|
54
|
Pio R, Senent Y, Tavira B, Ajona D. Fasting and fasting-mimicking conditions in the cancer immunotherapy era. J Physiol Biochem 2024:10.1007/s13105-024-01020-3. [PMID: 38587595 DOI: 10.1007/s13105-024-01020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
Fasting and fasting-mimicking conditions modulate tumor metabolism and remodel the tumor microenvironment (TME), which could be exploited for the treatment of tumors. A body of evidence demonstrates that fasting and fasting-mimicking conditions can kill cancer cells, or sensitize them to the antitumor activity of standard-of-care drugs while protecting normal cells against their toxic side effects. Pre- and clinical data also suggest that immune responses are involved in these therapeutic effects. Therefore, there is increasing interest in evaluating the impact of fasting-like conditions in the efficacy of antitumor therapies based on the restoration or activation of antitumor immune responses. Here, we review the recent progress in the intersection of fasting-like conditions and current cancer treatments, with an emphasis on cancer immunotherapy.
Collapse
Affiliation(s)
- Ruben Pio
- Laboratory of Translational Oncology, Program in Solid Tumors, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Yaiza Senent
- Laboratory of Translational Oncology, Program in Solid Tumors, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Beatriz Tavira
- Laboratory of Translational Oncology, Program in Solid Tumors, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Daniel Ajona
- Laboratory of Translational Oncology, Program in Solid Tumors, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain.
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain.
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
55
|
Jeong M, Collins N. Nutritional modulation of antitumor immunity. Curr Opin Immunol 2024; 87:102422. [PMID: 38728931 PMCID: PMC11288377 DOI: 10.1016/j.coi.2024.102422] [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: 03/15/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024]
Abstract
The composition and quantity of food we eat have a drastic impact on the development and function of immune responses. In this review, we highlight defined nutritional interventions shown to enhance antitumor immunity, including ketogenic, low-protein, high-fructose, and high-fiber diets, as well as dietary restriction. We propose that incorporating such nutritional interventions into immunotherapy protocols has the potential to increase therapeutic responsiveness and long-term tumor control in patients with cancer.
Collapse
Affiliation(s)
- Mingeum Jeong
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Nicholas Collins
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|
56
|
Heath H, Mogol AN, Santaliz Casiano A, Zuo Q, Madak-Erdogan Z. Targeting systemic and gut microbial metabolism in ER + breast cancer. Trends Endocrinol Metab 2024; 35:321-330. [PMID: 38220576 DOI: 10.1016/j.tem.2023.12.008] [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: 09/15/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
Estrogen receptor-positive (ER+) breast tumors have a better overall prognosis than ER- tumors; however, there is a sustained risk of recurrence. Mounting evidence indicates that genetic and epigenetic changes associated with resistance impact critical signaling pathways governing cell metabolism. This review delves into recent literature concerning the metabolic pathways regulated in ER+ breast tumors by the availability of nutrients and endocrine therapies and summarizes research on how changes in systemic and gut microbial metabolism can affect ER activity and responsiveness to endocrine therapy. As targeting of metabolic pathways using dietary or pharmacological approaches enters the clinic, we provide an overview of the supporting literature and suggest future directions.
Collapse
Affiliation(s)
- Hannah Heath
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Ayca Nazli Mogol
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Qianying Zuo
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
57
|
Raucci F, Vernieri C, Di Tano M, Ligorio F, Blaževitš O, Lazzeri S, Shmahala A, Fragale G, Salvadori G, Varano G, Casola S, Buono R, Visco E, de Braud F, Longo VD. Cyclic Fasting-Mimicking Diet Plus Bortezomib and Rituximab Is an Effective Treatment for Chronic Lymphocytic Leukemia. Cancer Res 2024; 84:1133-1148. [PMID: 38241703 PMCID: PMC10982641 DOI: 10.1158/0008-5472.can-23-0295] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 10/12/2023] [Accepted: 01/18/2024] [Indexed: 01/21/2024]
Abstract
Cyclic fasting-mimicking diet (FMD) is an experimental nutritional intervention with potent antitumor activity in preclinical models of solid malignancies. FMD cycles are also safe and active metabolically and immunologically in cancer patients. Here, we reported on the outcome of FMD cycles in two patients with chronic lymphocytic leukemia (CLL) and investigated the effects of fasting and FMD cycles in preclinical CLL models. Fasting-mimicking conditions in murine CLL models had mild cytotoxic effects, which resulted in apoptosis activation mediated in part by lowered insulin and IGF1 concentrations. In CLL cells, fasting conditions promoted an increase in proteasome activity that served as a starvation escape pathway. Pharmacologic inhibition of this escape mechanism with the proteasome inhibitor bortezomib resulted in a strong enhancement of the proapoptotic effects of starvation conditions in vitro. In mouse CLL models, combining cyclic fasting/FMD with bortezomib and rituximab, an anti-CD20 antibody, delayed CLL progression and resulted in significant prolongation of mouse survival. Overall, the effect of proteasome inhibition in combination with FMD cycles in promoting CLL death supports the targeting of starvation escape pathways as an effective treatment strategy that should be tested in clinical trials. SIGNIFICANCE Chronic lymphocytic leukemia cells resist fasting-mimicking diet by inducing proteasome activation to escape starvation, which can be targeted using proteasome inhibition by bortezomib treatment to impede leukemia progression and prolong survival.
Collapse
Affiliation(s)
- Franca Raucci
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Claudio Vernieri
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maira Di Tano
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Weill Cornell Medical College, Department of Medicine, Cornell University, New York, New York
| | - Francesca Ligorio
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Olga Blaževitš
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Samuel Lazzeri
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Giuseppe Fragale
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giulia Salvadori
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Gabriele Varano
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Stefano Casola
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Roberta Buono
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
- Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, California
| | - Euplio Visco
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valter D. Longo
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, California
| |
Collapse
|
58
|
Xiao YL, Gong Y, Qi YJ, Shao ZM, Jiang YZ. Effects of dietary intervention on human diseases: molecular mechanisms and therapeutic potential. Signal Transduct Target Ther 2024; 9:59. [PMID: 38462638 PMCID: PMC10925609 DOI: 10.1038/s41392-024-01771-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Diet, serving as a vital source of nutrients, exerts a profound influence on human health and disease progression. Recently, dietary interventions have emerged as promising adjunctive treatment strategies not only for cancer but also for neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, and metabolic disorders. These interventions have demonstrated substantial potential in modulating metabolism, disease trajectory, and therapeutic responses. Metabolic reprogramming is a hallmark of malignant progression, and a deeper understanding of this phenomenon in tumors and its effects on immune regulation is a significant challenge that impedes cancer eradication. Dietary intake, as a key environmental factor, can influence tumor metabolism. Emerging evidence indicates that dietary interventions might affect the nutrient availability in tumors, thereby increasing the efficacy of cancer treatments. However, the intricate interplay between dietary interventions and the pathogenesis of cancer and other diseases is complex. Despite encouraging results, the mechanisms underlying diet-based therapeutic strategies remain largely unexplored, often resulting in underutilization in disease management. In this review, we aim to illuminate the potential effects of various dietary interventions, including calorie restriction, fasting-mimicking diet, ketogenic diet, protein restriction diet, high-salt diet, high-fat diet, and high-fiber diet, on cancer and the aforementioned diseases. We explore the multifaceted impacts of these dietary interventions, encompassing their immunomodulatory effects, other biological impacts, and underlying molecular mechanisms. This review offers valuable insights into the potential application of these dietary interventions as adjunctive therapies in disease management.
Collapse
Affiliation(s)
- Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yue Gong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
59
|
Xie J, Deng X, Xie Y, Zhu H, Liu P, Deng W, Ning L, Tang Y, Sun Y, Tang H, Cai M, Xie X, Zou Y. Multi-omics analysis of disulfidptosis regulators and therapeutic potential reveals glycogen synthase 1 as a disulfidptosis triggering target for triple-negative breast cancer. MedComm (Beijing) 2024; 5:e502. [PMID: 38420162 PMCID: PMC10901283 DOI: 10.1002/mco2.502] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Disruption of disulfide homeostasis during biological processes can have fatal consequences. Excess disulfides induce cell death in a novel manner, termed as "disulfidptosis." However, the specific mechanism of disulfidptosis has not yet been elucidated. To determine the cancer types sensitive to disulfidptosis and outline the corresponding treatment strategies, we firstly investigated the crucial functions of disulfidptosis regulators pan-cancer at multi-omics levels. We found that different tumor types expressed dysregulated levels of disulfidptosis regulators, most of which had an impact on tumor prognosis. Moreover, we calculated the disulfidptosis activity score in tumors and validated it using multiple independent datasets. Additionally, we found that disulfidptosis activity was correlated with classic biological processes and pathways in various cancers. Disulfidptosis activity was also associated with tumor immune characteristics and could predict immunotherapy outcomes. Notably, the disulfidptosis regulator, glycogen synthase 1 (GYS1), was identified as a promising target for triple-negative breast cancer and validated via in vitro and in vivo experiments. In conclusion, our study elucidated the complex molecular phenotypes and clinicopathological correlations of disulfidptosis regulators in tumors, laying a solid foundation for the development of disulfidptosis-targeting strategies for cancer treatment.
Collapse
Affiliation(s)
- Jindong Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Xinpei Deng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Yi Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Hongbo Zhu
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - Peng Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Wei Deng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Li Ning
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Yuhui Tang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Yuying Sun
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Hailin Tang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Manbo Cai
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Yutian Zou
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| |
Collapse
|
60
|
Berrino F, Villarini A, Gargano G, Krogh V, Grioni S, Bellegotti M, Venturelli E, Raimondi M, Traina A, Zarcone M, Amodio R, Mano MP, Johansson H, Panico S, Santucci de Magistris M, Barbero M, Gavazza C, Mercandino A, Consolaro E, Galasso R, Del Riccio L, Bassi MC, Simeoni M, Premoli P, Pasanisi P, Bonanni B, Bruno E. The Effect of Diet on Breast Cancer Recurrence: The DIANA-5 Randomized Trial. Clin Cancer Res 2024; 30:965-974. [PMID: 37847493 PMCID: PMC10905522 DOI: 10.1158/1078-0432.ccr-23-1615] [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: 05/31/2023] [Revised: 08/02/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
PURPOSE The DIANA-5 randomized controlled trial assessed the effectiveness of a diet based on Mediterranean and macrobiotic traditions (macro-Mediterranean diet) in reducing breast cancer recurrence. PATIENTS AND METHODS The DIANA-5 study involved 1,542 patients with breast cancer at high risk of recurrence because of estrogen receptor-negative cancer, or metabolic syndrome, or high plasma levels of insulin or testosterone. Women were randomly assigned to an active dietary intervention (IG) or a control group (CG). Both groups received the 2007 American Institute for Cancer Research/World Cancer Research Fund recommendations for cancer prevention. The intervention consisted of meetings with kitchen classes, community meals, and dietary recommendations. Recommended foods included whole grain cereals, legumes, soy products, vegetables, fruit, nuts, olive oil, and fish. Foods to be avoided were refined products, potatoes, sugar and desserts, red and processed meat, dairy products, and alcoholic drinks. A compliance Dietary Index was defined by the difference between recommended and discouraged foods. RESULTS Over the 5 years of follow-up, 95 patients of the IG and 98 of the CG developed breast cancer recurrence [HR = 0.99; 95% confidence interval (CI): 0.69-1.40]. The analysis by compliance to the dietary recommendations (IG and CG together) showed that the women in the upper tertile of Dietary Index change had an HR of recurrence of 0.59 (95% CI: 0.36-0.92) compared with women in the lower tertile. CONCLUSIONS The DIANA-5 dietary intervention trial failed to show a reduction in breast cancer recurrence, although self-reported diet at year 1 in IG and CG combined showed a protective association with the higher Dietary Index change. See related commentary by McTiernan, p. 931.
Collapse
Affiliation(s)
- Franco Berrino
- Department of Epidemiology and Data Science, S.C. Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
- La Grande Via Foundation, Milan, Italy
| | - Anna Villarini
- Department of Medicine and Surgery, Section of Hygiene and Public Health, University of Perugia, Perugia, Italy
| | - Giuliana Gargano
- Department of Epidemiology and Data Science, S.C. Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Vittorio Krogh
- Department of Epidemiology and Data Science, S.C. Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Sara Grioni
- Department of Epidemiology and Data Science, S.C. Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Manuela Bellegotti
- Department of Epidemiology and Data Science, S.C. Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elisabetta Venturelli
- Department of Experimental Oncology, S.S.D. Nutrition Research and Metabolomics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Milena Raimondi
- Department of Epidemiology and Data Science, S.C. Epidemiology and Prevention, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Adele Traina
- Palermo and Province Cancer Registry, Clinical Epidemiology Unit with Cancer Registry, Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone”, University of Palermo, Palermo, Italy
| | - Maurizio Zarcone
- Palermo and Province Cancer Registry, Clinical Epidemiology Unit with Cancer Registry, Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone”, University of Palermo, Palermo, Italy
| | - Rosalba Amodio
- Palermo and Province Cancer Registry, Clinical Epidemiology Unit with Cancer Registry, Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone”, University of Palermo, Palermo, Italy
| | - Maria Piera Mano
- Dipartimento Scienze Chirurgiche, Study University, Turin, Italy
- S.C. Epidemiologia dei Tumori, AOU Città della Salute e della Scienza, CPO Piemonte, Turin, Italy
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | | | | | | | | | | | - Rocco Galasso
- Unit of Regional Cancer Registry, Clinical Epidemiology and Biostatistics, IRCCS-CROB, Rionero in Vulture, Italy
| | - Luciana Del Riccio
- Unit of Regional Cancer Registry, Clinical Epidemiology and Biostatistics, IRCCS-CROB, Rionero in Vulture, Italy
| | | | | | | | - Patrizia Pasanisi
- Department of Experimental Oncology, S.S.D. Nutrition Research and Metabolomics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Eleonora Bruno
- Department of Experimental Oncology, S.S.D. Nutrition Research and Metabolomics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
61
|
Giuliani G, Longo VD. Ketone bodies in cell physiology and cancer. Am J Physiol Cell Physiol 2024; 326:C948-C963. [PMID: 38189128 DOI: 10.1152/ajpcell.00441.2023] [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: 09/12/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
Ketogenic diets (KDs), fasting, or prolonged physical activity elevate serum ketone bodies (KBs) levels, providing an alternative fuel source for the brain and other organs. However, KBs play pleiotropic roles that go beyond their role in energy production. KBs can act as signaling metabolites, influence gene expression, proteins' posttranslational modifications (PTMs), inflammation, and oxidative stress. Here, we explore the impact of KBs on mammalian cell physiology, including aging and tissue regeneration. We also concentrate on KBs and cancer, given the extensive evidence that dietary approaches inducing ketosis, including fasting-mimicking diets (FMDs) and KDs, can prevent cancer and affect tumor progression.
Collapse
Affiliation(s)
- Giacomo Giuliani
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Valter D Longo
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
- IFOM, FIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
62
|
Menyhárt O, Győrffy B. Dietary approaches for exploiting metabolic vulnerabilities in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189062. [PMID: 38158024 DOI: 10.1016/j.bbcan.2023.189062] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Renewed interest in tumor metabolism sparked an enthusiasm for dietary interventions to prevent and treat cancer. Changes in diet impact circulating nutrient levels in the plasma and the tumor microenvironment, and preclinical studies suggest that dietary approaches, including caloric and nutrient restrictions, can modulate tumor initiation, progression, and metastasis. Cancers are heterogeneous in their metabolic dependencies and preferred energy sources and can be addicted to glucose, fructose, amino acids, or lipids for survival and growth. This dependence is influenced by tumor type, anatomical location, tissue of origin, aberrant signaling, and the microenvironment. This review summarizes nutrient dependencies and the related signaling pathway activations that provide targets for nutritional interventions. We examine popular dietary approaches used as adjuvants to anticancer therapies, encompassing caloric restrictions, including time-restricted feeding, intermittent fasting, fasting-mimicking diets (FMDs), and nutrient restrictions, notably the ketogenic diet. Despite promising results, much of the knowledge on dietary restrictions comes from in vitro and animal studies, which may not accurately reflect real-life situations. Further research is needed to determine the optimal duration, timing, safety, and efficacy of dietary restrictions for different cancers and treatments. In addition, well-designed human trials are necessary to establish the link between specific metabolic vulnerabilities and targeted dietary interventions. However, low patient compliance in clinical trials remains a significant challenge.
Collapse
Affiliation(s)
- Otília Menyhárt
- Semmelweis University, Department of Bioinformatics, Tűzoltó u. 7-9, H-1094 Budapest, Hungary; Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Balázs Győrffy
- Semmelweis University, Department of Bioinformatics, Tűzoltó u. 7-9, H-1094 Budapest, Hungary; Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117 Budapest, Hungary.
| |
Collapse
|
63
|
Bacci M, Lorito N, Smiriglia A, Subbiani A, Bonechi F, Comito G, Morriset L, El Botty R, Benelli M, López-Velazco JI, Caffarel MM, Urruticoechea A, Sflomos G, Malorni L, Corsini M, Ippolito L, Giannoni E, Meattini I, Matafora V, Havas K, Bachi A, Chiarugi P, Marangoni E, Morandi A. Acetyl-CoA carboxylase 1 controls a lipid droplet-peroxisome axis and is a vulnerability of endocrine-resistant ER + breast cancer. Sci Transl Med 2024; 16:eadf9874. [PMID: 38416843 DOI: 10.1126/scitranslmed.adf9874] [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: 11/24/2022] [Accepted: 01/30/2024] [Indexed: 03/01/2024]
Abstract
Targeting aromatase deprives ER+ breast cancers of estrogens and is an effective therapeutic approach for these tumors. However, drug resistance is an unmet clinical need. Lipidomic analysis of long-term estrogen-deprived (LTED) ER+ breast cancer cells, a model of aromatase inhibitor resistance, revealed enhanced intracellular lipid storage. Functional metabolic analysis showed that lipid droplets together with peroxisomes, which we showed to be enriched and active in the LTED cells, controlled redox homeostasis and conferred metabolic adaptability to the resistant tumors. This reprogramming was controlled by acetyl-CoA-carboxylase-1 (ACC1), whose targeting selectively impaired LTED survival. However, the addition of branched- and very long-chain fatty acids reverted ACC1 inhibition, a process that was mediated by peroxisome function and redox homeostasis. The therapeutic relevance of these findings was validated in aromatase inhibitor-treated patient-derived samples. Last, targeting ACC1 reduced tumor growth of resistant patient-derived xenografts, thus identifying a targetable hub to combat the acquisition of estrogen independence in ER+ breast cancers.
Collapse
Affiliation(s)
- Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Angela Subbiani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Francesca Bonechi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Ludivine Morriset
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France
| | - Matteo Benelli
- Department of Medical Oncology, Azienda USL Toscana Centro, Hospital of Prato, Via Suor Niccolina Infermiera 20, 59100 Prato, Italy
| | - Joanna I López-Velazco
- Biodonostia Health Research Institute, Paseo Dr Begiristain s/n, 20014 San Sebastian, Spain
| | - Maria M Caffarel
- Biodonostia Health Research Institute, Paseo Dr Begiristain s/n, 20014 San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Ander Urruticoechea
- Biodonostia Health Research Institute, Paseo Dr Begiristain s/n, 20014 San Sebastian, Spain
- Gipuzkoa Cancer Unit, OSI Donostialdea-Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014 San Sebastian, Spain
| | - George Sflomos
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Luca Malorni
- Department of Medical Oncology, Azienda USL Toscana Centro, Hospital of Prato, Via Suor Niccolina Infermiera 20, 59100 Prato, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, 25123 Brescia, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Vittoria Matafora
- IFOM ETS-AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Kristina Havas
- IFOM ETS-AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Angela Bachi
- IFOM ETS-AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
64
|
Mecca M, Picerno S, Cortellino S. The Killer's Web: Interconnection between Inflammation, Epigenetics and Nutrition in Cancer. Int J Mol Sci 2024; 25:2750. [PMID: 38473997 DOI: 10.3390/ijms25052750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Inflammation is a key contributor to both the initiation and progression of tumors, and it can be triggered by genetic instability within tumors, as well as by lifestyle and dietary factors. The inflammatory response plays a critical role in the genetic and epigenetic reprogramming of tumor cells, as well as in the cells that comprise the tumor microenvironment. Cells in the microenvironment acquire a phenotype that promotes immune evasion, progression, and metastasis. We will review the mechanisms and pathways involved in the interaction between tumors, inflammation, and nutrition, the limitations of current therapies, and discuss potential future therapeutic approaches.
Collapse
Affiliation(s)
- Marisabel Mecca
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Simona Picerno
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Salvatore Cortellino
- Laboratory of Preclinical and Translational Research, Responsible Research Hospital, 86100 Campobasso, CB, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, 80138 Naples, NA, Italy
- S.H.R.O. Italia Foundation ETS, 10060 Candiolo, TO, Italy
| |
Collapse
|
65
|
Fang G, Chen Q, Li J, Lian X, Shi D. The Diurnal Transcriptome Reveals the Reprogramming of Lung Adenocarcinoma Cells Under a Time-Restricted Feeding-Mimicking Regimen. J Nutr 2024; 154:354-368. [PMID: 38065409 DOI: 10.1016/j.tjnut.2023.11.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND The processes of tumor growth and circadian rhythm are intimately intertwined; thus, rewiring circadian metabolism by time-restricted feeding (TRF) may contribute to delaying carcinogenesis. However, research on the effect of a TRF cellular regimen on cancer is lacking. OBJECTIVE Investigate the circadian signatures of TRF in lung cancer in vitro. METHODS We first developed a cellular paradigm mimicking in vivo TRF and collected cells for transcriptome analysis. We further confirmed the effect on tumor cells upon 6-h TRF-mimicking (6-h TRFM) by real-time PCR, Lumicycle experiments, CCK-8, and flow cytometry assays. RESULTS We found that A549 lung adenocarcinoma cells treated with 6-h TRFM conditions displayed robust diurnal rhythms of transcriptomes, as well as modulation of the core clock genes relative to other different cellular regimens used in this study, including the fasting-mimicking conditions (ie, short-term starvation) and the serum-free regime. Notably, pathway analysis of oscillating genes exclusively in 6-h TRFM showed that some circadian genes were enriched in tumor-related pathways, such as the oxytocin signaling pathway, HIF-1 signaling pathway, and pentose and glucuronate interconversions. Moreover, in line with the circadian pathway enrichment results, 6-h TRFM robustly inhibited cell proliferation and induced cell apoptosis and cell cycle arrest in lung adenocarcinoma A549 cells, lung adenocarcinoma H460 cells, esophageal carcinoma Eca-109 cells, and breast adenocarcinoma MCF-7 cells. CONCLUSIONS Our findings provide the first in vitro mimicking medium for TRF intervention and indicate that 6-h TRFM is sufficient to reprogram the circadian signatures of lung adenocarcinoma cells and inhibit the progression of multiple tumors.
Collapse
Affiliation(s)
- Gaofeng Fang
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, P.R. China; Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Qianyao Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, P.R. China; Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Jianling Li
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, P.R. China; Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Xuemei Lian
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, P.R. China; Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, P.R. China.
| | - Dan Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, P.R. China; Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, P.R. China; Research Center for Environment and Population Health, School of Public Health, Chongqing Medical University, Chongqing, P.R. China.
| |
Collapse
|
66
|
Pfefferkorn RM, Mortzfeld BM, Fink C, von Frieling J, Bossen J, Esser D, Kaleta C, Rosenstiel P, Heine H, Roeder T. Recurrent Phases of Strict Protein Limitation Inhibit Tumor Growth and Restore Lifespan in A Drosophila Intestinal Cancer Model. Aging Dis 2024; 15:226-244. [PMID: 37962464 PMCID: PMC10796089 DOI: 10.14336/ad.2023.0517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 11/15/2023] Open
Abstract
Diets that restrict caloric or protein intake offer a variety of benefits, including decreasing the incidence of cancer. However, whether such diets pose a substantial therapeutic benefit as auxiliary cancer treatments remains unclear. We determined the effects of severe protein depletion on tumorigenesis in a Drosophila melanogaster intestinal tumor model, using a human RAF gain-of-function allele. Severe and continuous protein restriction significantly reduced tumor growth but resulted in premature death. Therefore, we developed a diet in which short periods of severe protein restriction alternated cyclically with periods of complete feeding. This nutritional regime reduced tumor mass, restored gut functionality, and rescued the lifespan of oncogene-expressing flies to the levels observed in healthy flies on a continuous, fully nutritious diet. Furthermore, this diet reduced the chemotherapy-induced stem cell activity associated with tumor recurrence. Transcriptome analysis revealed long-lasting changes in the expression of key genes involved in multiple major developmental signaling pathways. Overall, the data suggest that recurrent severe protein depletion effectively mimics the health benefits of continuous protein restriction, without undesired nutritional shortcomings. This provides seminal insights into the mechanisms of the memory effect required to maintain the positive effects of protein restriction throughout the phases of a full diet. Finally, the repetitive form of strict protein restriction is an ideal strategy for adjuvant cancer therapy that is useful in many tumor contexts.
Collapse
Affiliation(s)
- Roxana M. Pfefferkorn
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Benedikt M. Mortzfeld
- Department of Cell and Developmental Biology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Christine Fink
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Jakob von Frieling
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Judith Bossen
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Daniela Esser
- Department of Neuroimmunology, Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel, Germany.
| | - Christoph Kaleta
- Department Medical Systems Biology, Institute for Experimental Medicine, Kiel University, Germany.
| | - Philip Rosenstiel
- Department Molecular Cell Biology, Institute for Clinical Molecular Biology, Kiel University, Germany.
| | - Holger Heine
- Division of Innate Immunity, Research Center Borstel - Leibniz Lung Center, Borstel, Germany.
| | - Thomas Roeder
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| |
Collapse
|
67
|
Lin X, Gao Y. A bibliometric analysis of the Fasting-Mimicking Diet. Front Nutr 2024; 11:1328450. [PMID: 38321992 PMCID: PMC10844425 DOI: 10.3389/fnut.2024.1328450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/08/2024] Open
Abstract
The Fasting-Mimicking Diet (FMD) is a nutritional strategy that involves significantly reducing calorie intake for a specific period to mimic the physiological effects of fasting while still providing the body with nutrition. Our study aimed to conduct a bibliometric study to explore the latest publishing trends and areas of intense activity within the sphere of FMD. We extracted data on FMD publications from the Web of Science Core Collection (WOSCC) database. The bibliometric analysis was conducted by WOSCC Online Analysis Platform and VOSviewer 1.6.16. In total, there were 169 publications by 945 authors from 342 organizations and 25 countries/regions, and published in 111 journals. The most productive country, organization, author, and journal were the United States, the University of Southern California, Valter D. Longo, and Nutrients, respectively. The first high-cited document was published in Ageing Research Reviews and authored by Mattson et al. In this study, they discuss the various health benefits of FMD including improved metabolic health, weight management, and even potential effects on delaying aging processes and reducing the risk of chronic diseases. In conclusion, our study is the first bibliometric analysis of the FMD. The main research hotspots and frontiers were FMD for cancer, FMD for metabolic-related diseases, and FMD for cognitive improvement. FMD may have some potential benefits for multiple diseases which should be further investigated.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Hangzhou, Zhejiang, China
| | - Yue Gao
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Hangzhou, Zhejiang, China
| |
Collapse
|
68
|
Lin X, Wang S, Huang J. The effects of time-restricted eating for patients with nonalcoholic fatty liver disease: a systematic review. Front Nutr 2024; 10:1307736. [PMID: 38239843 PMCID: PMC10794638 DOI: 10.3389/fnut.2023.1307736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a significant global health concern. Numerous investigations have explored the implications of time-restricted eating (TRE) in the management of NAFLD. Therefore, the objective of our study was to conduct a systematic review to summarize and analyze all randomized controlled trials (RCTs) of TRE for patients with NAFLD. A thorough literature search was executed across Embase, Cochrane Library, and PubMed databases, covering all records from their inception until 1 September 2023. All clinical studies of TRE for NAFLD were summarized and analyzed. Our systematic review included four RCTs, encompassing a total of 443 NAFLD patients. These studies varied in sample size from 32 to 271 participants. The TRE intervention was consistently applied in an 8-h window, over durations ranging from 4 weeks to 12 months. The findings suggest that TRE could offer several health benefits for NAFLD patients, such as improved liver health indicators like liver stiffness and intrahepatic triglyceride (IHTG) levels. Consequently, TRE appears to be a promising dietary intervention for NAFLD patients. However, it is premature to recommend TRE for patients with NAFLD. The existing body of research on the effects of TRE in NAFLD contexts is limited, underscoring the need for further high-quality studies to expand our understanding of TRE's benefits in treating NAFLD. Ongoing clinical trials may provide more insights into the effects of TRE in NAFLD.
Collapse
Affiliation(s)
| | - Shuai Wang
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinyu Huang
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
69
|
Xiao B, Jiang Y, Yuan S, Cai L, Xu T, Jia L. Silibinin, a potential fasting mimetic, inhibits hepatocellular carcinoma by triggering extrinsic apoptosis. MedComm (Beijing) 2024; 5:e457. [PMID: 38222315 PMCID: PMC10784426 DOI: 10.1002/mco2.457] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 01/16/2024] Open
Abstract
Fasting, without inducing malnutrition, has been shown to have various beneficial effects, including the inhibition of tumor initiation and progression. However, prolonged fasting poses challenges for many cancer patients, particularly those in intermediate and terminal stages. Thus, there is an urgent need for the development of fasting mimetics which harness the protective effects of fasting but more suitable for patients. In this study, we first highlighted the pivotal role of silibinin in AMP-activated protein kinase (AMPK) pathway and may serve, as a potential fasting mimetic via screening hepatoprotective drugs. Further metabolic analysis showed that silibinin inhibited the adenosine triphosphate (ATP) levels, glucose uptake and diminished glycolysis process, which further confirmed that silibinin served as a fasting mimetic. In addition, fasting synergized with silibinin, or used independently, to suppress the growth of hepatocellular carcinoma (HCC) in vivo. Mechanistically, silibinin upregulated death receptor 5 (DR5) through AMPK activation, and thus promoting extrinsic apoptosis and inhibiting HCC growth both in vitro and in vivo. Inhibition of AMPK using small interfering RNA (siRNA) or compound C, an AMPK inhibitor, significantly attenuated the upregulation of DR5 and the apoptotic response induced by silibinin. These findings suggest that silibinin holds promise as a fasting mimetic and may serve as an adjuvant drug for HCC treatment.
Collapse
Affiliation(s)
- Biying Xiao
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yanyu Jiang
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shuying Yuan
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lili Cai
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tong Xu
- Departmnent of OncologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Lijun Jia
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
70
|
Ligorio F, Lobefaro R, Fucà G, Provenzano L, Zanenga L, Nasca V, Sposetti C, Salvadori G, Ficchì A, Franza A, Martinetti A, Sottotetti E, Formisano B, Depretto C, Scaperrotta G, Belfiore A, Vingiani A, Ferraris C, Pruneri G, de Braud F, Vernieri C. Adding fasting-mimicking diet to first-line carboplatin-based chemotherapy is associated with better overall survival in advanced triple-negative breast cancer patients: A subanalysis of the NCT03340935 trial. Int J Cancer 2024; 154:114-123. [PMID: 37615485 DOI: 10.1002/ijc.34701] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023]
Abstract
Severe calorie restriction, in the form of cyclic fasting or fasting-mimicking diets (FMDs), boosts the antitumor activity of cytotoxic chemotherapy in mouse models of triple-negative breast cancer (TNBC). This effect is mostly mediated by fasting/FMD-induced reduction of plasma glucose concentration and by a boost in antitumor immunity. However, clinical evidence that cyclic FMD may impact on the outcomes of advanced TNBC (aTNBC) patients is lacking. We compared the overall survival (OS) of 14 aTNBC patients receiving first-line carboplatin-gemcitabine plus cyclic FMD in the context of the NCT03340935 trial with the OS of 76 consecutive aTNBC patients treated with carboplatin-based chemotherapy alone at Fondazione IRCCS Istituto Nazionale dei Tumori. Multivariable Cox regression models were used to adjust the prognostic impact of FMD for other prognostic variables. Patients undergoing cyclic FMD in combination with carboplatin-gemcitabine had better OS when compared to patients receiving chemotherapy alone (median OS 30.3 months, 95% CI 18-NR, vs 17.2 months, 95% CI 15.3-25.1, log-rank P value .041). Multivariable analysis confirmed an association between FMD use and better OS (HR: 0.40; 95% CI: 0.19-0.86; P = .019) also after propensity score-based matching according to patient ECOG PS and the presence of de novo metastatic disease (HR: 0.41; 95% CI: 0.21-0.83; P = .013). Cyclic FMD in combination with first-line chemotherapy may improve clinical outcomes in aTNBC patients. Our study paves the way for conducting phase II trials to investigate if cyclic FMD can increase the antitumor activity/efficacy of chemotherapy or chemoimmunotherapy in patients with early-stage TNBC or aTNBC.
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Lucrezia Zanenga
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vincenzo Nasca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Caterina Sposetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Giulia Salvadori
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Angela Ficchì
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Franza
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Formisano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Catherine Depretto
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Antonino Belfiore
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Vingiani
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Cristina Ferraris
- Breast Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giancarlo Pruneri
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
71
|
Haif SK, Al Kury LT, Talib WH. Combination of Thymoquinone and Intermittent Fasting as a Treatment for Breast Cancer Implanted in Mice. PLANTS (BASEL, SWITZERLAND) 2023; 13:35. [PMID: 38202341 PMCID: PMC10780740 DOI: 10.3390/plants13010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
Breast cancer stands out as a particularly challenging form of cancer to treat among various types. Traditional treatment methods have been longstanding approaches, yet their efficacy has diminished over time owing to heightened toxicity, adverse effects, and the emergence of multi-drug resistance. Nevertheless, a viable solution has emerged through the adoption of a complementary treatment strategy utilizing natural substances and the incorporation of intermittent fasting to enhance therapeutic outcomes. This study aimed to assess the anticancer activity of thymoquinone (TQ), intermittent fasting, and their combination using in vivo and in vitro methods. The anti-proliferative activity of TQ and fasting (glucose/serum restriction) were evaluated against the T47D, MDA-MB-231, and EMT6 cell lines and compared to normal cell lines (Vero) using the MTT colorimetric assay method. Additionally, this study aimed to determine the half-maximal inhibitory concentration (IC50) of TQ. For the in vivo experiment, the antitumor activity of TQ and intermittent fasting (IF) was assessed by measuring the tumor sizes using a digital caliper to determine the change in the tumor size and survival rates. At the molecular level, the serum levels of glucose, β-hydroxybutyrate (β-HB), leptin, and insulin growth factor-1 (IGF-1) were measured using standard kits. Additionally, the aspartate transaminase (AST), alanine transaminase (ALT), and creatinine serum levels were measured. The inhibition of the breast cancer cell lines was achieved by TQ. TQ and intermittent fasting both had an additional anticancer effect against breast tumors inoculated in mice. The combination therapy was evaluated and found to significantly reduce the tumor size, with a change in tumor size of -57.7%. Additionally, the combination of TQ and IF led to a decrease in the serum levels of glucose, IGF-1 (24.49 ng/mL) and leptin (1.77 ng/mL) while increasing β-hydroxybutyrate in the mice given combination therapy (200.86 nM) with no toxicity on the liver or kidneys. In the mice receiving combination therapy, TQ and IF treated breast cancer in an additive way without causing liver or kidney toxicity due to decreased levels of glucose, IGF-1, and leptin and increased levels of β-hydroxybutyrate. Further investigation is required to optimize the doses and determine the other possible mechanisms exhibited by the novel combination.
Collapse
Affiliation(s)
- Shatha Khaled Haif
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931-166, Jordan;
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates
| | - Wamidh H. Talib
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman 11931-166, Jordan
| |
Collapse
|
72
|
Buono R, Tucci J, Cutri R, Guidi N, Mangul S, Raucci F, Pellegrini M, Mittelman SD, Longo VD. Fasting-Mimicking Diet Inhibits Autophagy and Synergizes with Chemotherapy to Promote T-Cell-Dependent Leukemia-Free Survival. Cancers (Basel) 2023; 15:5870. [PMID: 38136414 PMCID: PMC10741737 DOI: 10.3390/cancers15245870] [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: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Fasting mimicking diets (FMDs) are effective in the treatment of many solid tumors in mouse models, but their effect on hematologic malignancies is poorly understood, particularly in combination with standard therapies. Here we show that cycles of a 3-day FMD given to high-fat-diet-fed mice once a week increased the efficacy of vincristine to improve survival from BCR-ABL B acute lymphoblastic leukemia (ALL). In mice fed a standard diet, FMD cycles in combination with vincristine promoted cancer-free survival. RNA seq and protein assays revealed a vincristine-dependent decrease in the expression of multiple autophagy markers, which was exacerbated by the fasting/FMD conditions. The autophagy inhibitor chloroquine could substitute for fasting/FMD to promote cancer-free survival in combination with vincristine. In vitro, targeted inhibition of autophagy genes ULK1 and ATG9a strongly potentiated vincristine's toxicity. Moreover, anti-CD8 antibodies reversed the effects of vincristine plus fasting/FMD in promoting leukemia-free survival in mice, indicating a central role of the immune system in this response. Thus, the inhibition of autophagy and enhancement of immune responses appear to be mediators of the fasting/FMD-dependent cancer-free survival in ALL mice.
Collapse
Affiliation(s)
- Roberta Buono
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Jonathan Tucci
- Center for Endocrinology, Diabetes & Metabolism, Children’s Hospital Los Angeles, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Raffaello Cutri
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
| | - Novella Guidi
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
| | - Serghei Mangul
- Department of Computer Science, University of California Los Angeles, 580 Portola Plaza, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, Boyer Hall, 611 Charles Young Drive, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Franca Raucci
- IFOM AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Matteo Pellegrini
- Institute for Quantitative and Computational Biosciences, Boyer Hall, 611 Charles Young Drive, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, 801 Hilgard Avenue, Los Angeles, CA 90095, USA
| | - Steven D. Mittelman
- Center for Endocrinology, Diabetes & Metabolism, Children’s Hospital Los Angeles, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
- Division of Pediatric Endocrinology, UCLA Mattel Children’s Hospital, 10833 Le Conte Avenue, MDCC 22-315, Los Angeles, CA 90095, USA
| | - Valter D. Longo
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
- IFOM AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
73
|
Zhang X, Zhong Y, Liu L, Jia C, Cai H, Yang J, Wu B, Lv Z. Fasting regulates mitochondrial function through lncRNA PRKCQ-AS1-mediated IGF2BPs in papillary thyroid carcinoma. Cell Death Dis 2023; 14:827. [PMID: 38092752 PMCID: PMC10719255 DOI: 10.1038/s41419-023-06348-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 11/01/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023]
Abstract
Recurring evidence suggests that fasting has extensive antitumor effects in various cancers, including papillary thyroid carcinoma (PTC). However, the underlying mechanism of this relationship with PTC is unknown. In this study, we study the effect of fasting on glycolysis and mitochondrial function in PTC. We find that fasting impairs glycolysis and reduces mitochondrial dysfunction in vitro and in vivo and also fasting in vitro and fasting mimicking diets (FMD) in vivo significantly increase the expression of lncRNA-protein kinase C theta antisense RNA 1 (PRKCQ-AS1), during the inhibition of TPC cell glycolysis and mitochondrial function. Moreover, lncRNA PRKCQ-AS1 was significantly lower in PTC tissues and cells. In addition, PRKCQ-AS1 overexpression increased PTC cell glycolysis and mitochondrial function; PRKCQ-AS1 knockdown has the opposite effect. On further mechanistic analysis, we identified that PRKCQ-AS1 physically interacts with IGF2BPs and enhances protein arginine methyltransferases 7 (PRMT7) mRNA, which is the key player in regulating glycolysis and mitochondrial function in PTC. Hence, PRKCQ-AS1 inhibits tumor growth while regulating glycolysis and mitochondrial functions via IGF2BPs/PRMT7 signaling. These results indicate that lncRNA PRKCQ-AS1 is a key downstream target of fasting and is involved in PTC metabolic reprogramming. Further, the PRKCQ-AS1/IGF2BPs/PRMT7 axis is an ideal therapeutic target for PTC diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai hospital Affiliated with Jinan University, Jinan University, 519000, Guangdong, China.
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China.
| | - Yong Zhong
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Lin Liu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Chengyou Jia
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Haidong Cai
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Jianshe Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Bo Wu
- Center of Thyroid, Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China.
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China.
| |
Collapse
|
74
|
Kondo S, Miyake M. Simultaneous Prediction Method for Intestinal Absorption and Metabolism Using the Mini-Ussing Chamber System. Pharmaceutics 2023; 15:2732. [PMID: 38140073 PMCID: PMC10747201 DOI: 10.3390/pharmaceutics15122732] [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: 10/12/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Many evaluation tools for predicting human absorption are well-known for using cultured cell lines such as Caco-2, MDCK, and so on. Since the combinatorial chemistry and high throughput screening system, pharmacological assay, and pharmaceutical profiling assay are mainstays of drug development, PAMPA has been used to evaluate human drug absorption. In addition, cultured cell lines from iPS cells have been attracting attention because they morphologically resemble human intestinal tissues. In this review, we used human intestinal tissues to estimate human intestinal absorption and metabolism. The Ussing chamber uses human intestinal tissues to directly assay a drug candidate's permeability and determine the electrophysiological parameters such as potential differences (PD), short circuit current (Isc), and resistance (R). Thus, it is an attractive tool for elucidating human intestinal permeability and metabolism. We have presented a novel prediction method for intestinal absorption and metabolism by utilizing a mini-Ussing chamber using human intestinal tissues and animal intestinal tissues, based on the transport index (TI). The TI value was calculated by taking the change in drug concentrations on the apical side due to precipitation and the total amounts accumulated in the tissue (Tcorr) and transported to the basal side (Xcorr). The drug absorbability in rank order, as well as the fraction of dose absorbed (Fa) in humans, was predicted, and the intestinal metabolism of dogs and rats was also predicted, although it was not quantitative. However, the metabolites formation index (MFI) values, which are included in the TI values, can predict the evaluation of intestinal metabolism and absorption by using ketoconazole. Therefore, the mini-Ussing chamber, equipped with human and animal intestinal tissues, would be an ultimate method to predict intestinal absorption and metabolism simultaneously.
Collapse
Affiliation(s)
- Satoshi Kondo
- Department of Drug Metabolism and Pharmacokinetics, Nonclinical Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., 460-10 Kagasuno Kawauchi-cho, Tokushima 771-0192, Japan;
- Department of Drug Safety Research, Nonclinical Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., 460-10 Kagasuno Kawauchi-cho, Tokushima 771-0192, Japan
| | - Masateru Miyake
- Business Integrity and External Affairs, Otsuka Pharmaceutical Co., Ltd., 2-16-4 Konan, Minato-ku, Tokyo 108-8242, Japan
| |
Collapse
|
75
|
Han SJ, Stacy A, Corral D, Link VM, De Siqueira MK, Chi L, Teijeiro A, Yong DS, Perez-Chaparro PJ, Bouladoux N, Lim AI, Enamorado M, Belkaid Y, Collins N. Microbiota configuration determines nutritional immune optimization. Proc Natl Acad Sci U S A 2023; 120:e2304905120. [PMID: 38011570 PMCID: PMC10710091 DOI: 10.1073/pnas.2304905120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/25/2023] [Indexed: 11/29/2023] Open
Abstract
Mild or transient dietary restriction (DR) improves many aspects of health and aging. Emerging evidence from us and others has demonstrated that DR also optimizes the development and quality of immune responses. However, the factors and mechanisms involved remain to be elucidated. Here, we propose that DR-induced optimization of immunological memory requires a complex cascade of events involving memory T cells, the intestinal microbiota, and myeloid cells. Our findings suggest that DR enhances the ability of memory T cells to recruit and activate myeloid cells in the context of a secondary infection. Concomitantly, DR promotes the expansion of commensal Bifidobacteria within the large intestine, which produce the short-chain fatty acid acetate. Acetate conditioning of the myeloid compartment during DR enhances the capacity of these cells to kill pathogens. Enhanced host protection during DR is compromised when Bifidobacteria expansion is prevented, indicating that microbiota configuration and function play an important role in determining immune responsiveness to this dietary intervention. Altogether, our study supports the idea that DR induces both memory T cells and the gut microbiota to produce distinct factors that converge on myeloid cells to promote optimal pathogen control. These findings suggest that nutritional cues can promote adaptation and co-operation between multiple immune cells and the gut microbiota, which synergize to optimize immunity and protect the collective metaorganism.
Collapse
Affiliation(s)
- Seong-Ji Han
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Apollo Stacy
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Dan Corral
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Verena M. Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | | | - Liang Chi
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Ana Teijeiro
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Daniel S. Yong
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - P. Juliana Perez-Chaparro
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Michel Enamorado
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Nicholas Collins
- Metaorganism Immunity Section, Laboratory of Host Immunity and the Microbiome, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| |
Collapse
|
76
|
Ni C, Li J. Take metabolic heterogeneity into consideration when applying dietary interventions to cancer therapy: A review. Heliyon 2023; 9:e22814. [PMID: 38213585 PMCID: PMC10782175 DOI: 10.1016/j.heliyon.2023.e22814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024] Open
Abstract
In recent years, dietary interventions have attracted much attention in cancer therapy. Mechanistic studies suggest that dietary interventions can inhibit the progression of cancer through deprivation of essential metabolites, lowering the levels of protumor hormones, activation of anticancer immunity and synergistic effects with conventional anticancer therapies. The feasibility, safety and promising tumor outcomes have also been established in humans. However, the results from both preclinical and clinical studies are inconsistent or even conflicting, the reasons for which have not been extensively considered. In this review, we discuss the various heterogeneity, including dietary protocols, tissue of origin and cancer locations, spatial and temporal metabolic heterogeneity, and divergent combination treatment, that may affect the responses of different cancers to dietary interventions. Understanding this heterogeneity and taking them into consideration when applying dietary interventions to cancer therapy will allow us to deliver the right diet to the right patient at the right time to maximize compliance, safety and efficacy of conventional anticancer therapy and to improve the outcomes of patients with cancer.
Collapse
Affiliation(s)
- Chun Ni
- Department of General Surgery, Chong Gang General Hospital, 400016, Chongqing, China
| | - Jian Li
- Department of General Surgery, the Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, China
| |
Collapse
|
77
|
Nelson AB, Queathem ED, Puchalska P, Crawford PA. Metabolic Messengers: ketone bodies. Nat Metab 2023; 5:2062-2074. [PMID: 38092961 DOI: 10.1038/s42255-023-00935-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/20/2023] [Indexed: 12/21/2023]
Abstract
Prospective molecular targets and therapeutic applications for ketone body metabolism have increased exponentially in the past decade. Initially considered to be restricted in scope as liver-derived alternative fuel sources during periods of carbohydrate restriction or as toxic mediators during diabetic ketotic states, ketogenesis and ketone bodies modulate cellular homeostasis in multiple physiological states through a diversity of mechanisms. Selective signalling functions also complement the metabolic fates of the ketone bodies acetoacetate and D-β-hydroxybutyrate. Here we discuss recent discoveries revealing the pleiotropic roles of ketone bodies, their endogenous sourcing, signalling mechanisms and impact on target organs, and considerations for when they are either stimulated for endogenous production by diets or pharmacological agents or administered as exogenous wellness-promoting agents.
Collapse
Affiliation(s)
- Alisa B Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Eric D Queathem
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
78
|
Swaby A, Atallah A, Varol O, Cristea A, Quail DF. Lifestyle and host determinants of antitumor immunity and cancer health disparities. Trends Cancer 2023; 9:1019-1040. [PMID: 37718223 DOI: 10.1016/j.trecan.2023.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023]
Abstract
Lifestyle factors exert profound effects on host physiology and immunology. Disparities in cancer outcomes persist as a complex and multifaceted challenge, necessitating a comprehensive understanding of the interplay between host environment and antitumor immune responses. Determinants of health - such as obesity, diet, exercise, stress, or sleep disruption - have the potential for modification, yet some exert long-lasting effects and may challenge the notion of complete reversibility. Herein we review intersectional considerations of lifestyle immunity and the impact on tumor immunology and disparities in cancer outcomes, with a focus on obesity.
Collapse
Affiliation(s)
- Anikka Swaby
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Aline Atallah
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Ozgun Varol
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Alyssa Cristea
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada; Department of Physiology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
79
|
Mishra S, Persons PA, Lorenzo AM, Chaliki SS, Bersoux S. Time-Restricted Eating and Its Metabolic Benefits. J Clin Med 2023; 12:7007. [PMID: 38002621 PMCID: PMC10672223 DOI: 10.3390/jcm12227007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Newer management strategies are being evaluated to treat obesity, which continues to increase worldwide. After 12 h of fasting, the body switches from glucose to fat metabolism, regulating protein synthesis and autophagy. These cellular responses are central to the metabolic benefits of time-restricted eating (TRE), independent of calorie restriction and weight loss, and they have heightened interest in TRE regimens. Controversy remains, however, regarding the benefits of TRE regimens. We reviewed the current literature and concluded that TRE is equivalent to calorie restriction for weight loss and has positive effects for patients with diseases such as nonalcoholic fatty liver disease, cancer, and cardiovascular disease.
Collapse
Affiliation(s)
- Sneha Mishra
- Division of Community Internal Medicine Mayo Clinic, Scottsdale, AZ 85259, USA; (P.A.P.); (A.M.L.); (S.S.C.); (S.B.)
| | | | | | | | | |
Collapse
|
80
|
Zhang J, Li X, Yin Y, Cao G, Wang H. A Biodegradable Nucleotide Coordination Polymer for Enhanced NSCLC Therapy in Combination with Metabolic Modulation. Adv Healthc Mater 2023; 12:e2302187. [PMID: 37607115 DOI: 10.1002/adhm.202302187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/09/2023] [Indexed: 08/24/2023]
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in the treatment of non-small cell lung cancer (NSCLC) still face challenges of acquired resistance and non-negligible side effects. To overcome these limitations, a biodegradable coordination polymer using guanine deoxynucleotide and ferrous iron (dGNP) is developed for targeted delivery of EGFR-TKIs. dGNPs can efficiently target nucleoside transporters in tumor cells that are regulated by fasting-mimicking diet (FMD). Meanwhile, FMD can augment the therapeutic efficacy of EGFR-TKIs by suppressing EGFR tyrosine kinase phosphorylation and related downstream pathways. In vivo results demonstrate that EGFR-TKIs-laden dGNPs combined with FMD treatment exhibit superior antitumor efficacy and reduced side effect. This study provides an innovative approach to enhance the therapeutic efficacy of EGFR-TKIs through nucleotide nanocarrier and metabolic modulation.
Collapse
Affiliation(s)
- Jie Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yue Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Guoliang Cao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
81
|
Zhong Z, Zhang H, Nan K, Zhong J, Wu Q, Lu L, Yue Y, Zhang Z, Guo M, Wang Z, Xia J, Xing Y, Fu Y, Yu B, Zhou W, Sun X, Shen Y, Chen W, Zhang J, Zhang J, Ma D, Chu Y, Liu R, Miao C. Fasting-Mimicking Diet Drives Antitumor Immunity against Colorectal Cancer by Reducing IgA-Producing Cells. Cancer Res 2023; 83:3529-3543. [PMID: 37602826 PMCID: PMC10618736 DOI: 10.1158/0008-5472.can-23-0323] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/05/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
As a safe, feasible, and inexpensive dietary intervention, fasting-mimicking diet (FMD) exhibits excellent antitumor efficacy by regulating metabolism and boosting antitumor immunity. A better understanding of the specific mechanisms underlying the immunoregulatory functions of FMD could help improve and expand the clinical application of FMD-mediated immunotherapeutic strategies. In this study, we aimed to elucidate the role of metabolic reprogramming induced by FMD in activation of antitumor immunity against colorectal cancer. Single-cell RNA sequencing analysis of intratumoral immune cells revealed that tumor-infiltrating IgA+ B cells were significantly reduced by FMD treatment, leading to the activation of antitumor immunity and tumor regression in murine colorectal cancer models. Mechanistically, FMD delayed tumor growth by repressing B-cell class switching to IgA. Therefore, FMD-induced reduction of IgA+ B cells overcame the suppression of CD8+ T cells. The immunoregulatory and antitumor effects of FMD intervention were reversed by IgA+ B-cell transfer. Moreover, FMD boosted fatty acid oxidation (FAO) to trigger RUNX3 acetylation, thus inactivating Cα gene transcription and IgA class switching. IgA+ B-cell expansion was also impeded in patients placed on FMD, while B-cell expression of carnitine palmitoyl transferase 1A (CPT1A), the rate-limiting enzyme of FAO, was increased. Furthermore, CPT1A expression was negatively correlated with both IgA+ B cells and IgA secretion within colorectal cancer. Together, these results highlight that FMD holds great promise for treating colorectal cancer. Furthermore, the degree of IgA+ B cell infiltration and FAO-associated metabolic status are potential biomarkers for evaluating FMD efficacy. SIGNIFICANCE Metabolic reprogramming of B cells induced by fasting-mimicking diet suppresses IgA class switching and production to activate antitumor immunity and inhibit tumor growth. See related commentary by Bush and Perry, p. 3493.
Collapse
Affiliation(s)
- Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Qichao Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Lihong Lu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhenyu Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Miaomiao Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, 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
| | - Jie Xia
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yun Xing
- 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
| | - 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
| | - Wenchang Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xingfeng Sun
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jie Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, 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, Shanghai, China
| |
Collapse
|
82
|
Burkett WC, Clontz AD, Keku TO, Bae-Jump V. The interplay of obesity, microbiome dynamics, and innovative anti-obesity strategies in the context of endometrial cancer progression and therapeutic approaches. Biochim Biophys Acta Rev Cancer 2023; 1878:189000. [PMID: 37844671 PMCID: PMC12046527 DOI: 10.1016/j.bbcan.2023.189000] [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: 04/30/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy in the United States, and its incidence and mortality are rising. Obesity is more tightly associated with EC than any other cancer. Thus, the rising prevalence of obesity and associated risk factors, including diabetes and insulin resistance, cause alarm. The metabolic derangements of obesity increase the bioavailability of estrogen, hyperinsulinemia, and inflammation in a complex system with direct and indirect effects on the endometrium, resulting in proliferation and, ultimately, carcinogenesis. In addition, the gut dysbiosis associated with obesity helps contribute to these metabolic derangements, priming an individual for developing EC and perhaps affecting treatment efficacy. More recent studies are beginning to explore obesity's effect on the local tumor microbiome of EC and its role in carcinogenesis. Significant and sustained weight loss in individuals can considerably decrease the risk of EC, likely through reversal of the altered metabolism and dysbiosis resulting obesity. Bariatric surgery is the gold standard for successful weight loss and highlights how reversing of the systemic effects of obesity can reduce EC risk. However, the current limited availability, knowledge, and imposed stigma of bariatric surgery prohibits population-level reductions in EC. Therefore, effective and maintainable non-surgical dietary and pharmacologic interventions are needed.
Collapse
Affiliation(s)
- Wesley C Burkett
- University of North Carolina at Chapel Hill, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, United States of America.
| | - Angela D Clontz
- University of North Carolina at Chapel Hill, Nutrition Research Institute, United States of America.
| | - Temitope O Keku
- University of North Carolina at Chapel Hill, Department of Medicine, Center for Gastrointestinal Biology and Disease, United States of America.
| | - Victoria Bae-Jump
- University of North Carolina at Chapel Hill, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, United States of America; University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, United States of America.
| |
Collapse
|
83
|
Noll JH, Levine BL, June CH, Fraietta JA. Beyond youth: Understanding CAR T cell fitness in the context of immunological aging. Semin Immunol 2023; 70:101840. [PMID: 37729825 DOI: 10.1016/j.smim.2023.101840] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
Population aging, a pervasive global demographic trend, is anticipated to challenge health and social systems worldwide. This phenomenon is due to medical advancements enabling longer lifespans, with 20% of the US population soon to be over 65 years old. Consequently, there will be a surge in age-related diseases. Senescence, characterized by the loss of biological maintenance and homeostasis at molecular and cellular levels, either correlates with or directly causes age-related phenotypic changes. Decline of the immune system is a critical factor in the senescence process, with cancer being a primary cause of death in elderly populations. Chimeric antigen receptor (CAR) T cell therapy, an innovative approach, has demonstrated success mainly in pediatric and young adult hematological malignancies but remains largely ineffective for diseases affecting older populations, such as late-in-life B cell malignancies and most solid tumor indications. This limitation arises because CAR T cell efficacy heavily relies on the fitness of the patient-derived starting T cell material. Numerous studies suggest that T cell senescence may be a key driver of CAR T cell deficiency. This review examines correlates and underlying factors associated with favorable CAR T cell outcomes and explores potential experimental and clinically actionable strategies for T cell rejuvenation.
Collapse
Affiliation(s)
- Julia Han Noll
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
84
|
Ligorio F, Provenzano L, Vernieri C. Fasting-mimicking diet: a metabolic approach for the treatment of breast cancer. Curr Opin Oncol 2023; 35:491-499. [PMID: 37621169 DOI: 10.1097/cco.0000000000000986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
PURPOSE OF REVIEW Metabolic reprogramming is a new and potentially targetable hallmark of cancer. In recent years, fasting and fasting-mimicking diets (FMDs) have been tested as anticancer strategies both in preclinical experiments and in clinical trials. In this review, we aim at summarizing the available evidence about the antitumour activity of these approaches in preclinical breast cancer models, as well as results from clinical trials investigating fasting/FMD in breast cancer patients. RECENT FINDINGS Preclinical evidence demonstrated that nutrient deprivation boosts the antitumor activity of chemotherapy, immunotherapy or targeted therapies in triple-negative breast cancer (TNBC) and HR+/HER2 models through both cell-autonomous antitumour effects in cancer cells and favourable modifications in intratumor immune cells. Several clinical experiences demonstrated that fasting/FMD is feasible and well tolerated in combination with standard treatments in BC patients, and that it could reduce chemotherapy-related toxicities. Finally, despite the absence of randomized trials demonstrating the antitumor activity of fasting/FMD in breast cancer patients, preliminary clinical reports suggest that this experimental nutritional strategy may enhance chemotherapy activity. Randomized clinical trials are ongoing to validate these results at a larger scale. SUMMARY Fasting/FMD is a promising therapeutic approach in patients with breast cancer; ongoing and future trials will confirm their role in improving breast cancer care.
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori
- Oncology and Hemato-Oncology Department, University of Milan
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori
- Oncology and Hemato-Oncology Department, University of Milan
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
85
|
Khalifa A, Guijarro A, Ravera S, Bertola N, Adorni MP, Papotti B, Raffaghello L, Benelli R, Becherini P, Namatalla A, Verzola D, Reverberi D, Monacelli F, Cea M, Pisciotta L, Bernini F, Caffa I, Nencioni A. Cyclic fasting bolsters cholesterol biosynthesis inhibitors' anticancer activity. Nat Commun 2023; 14:6951. [PMID: 37907500 PMCID: PMC10618279 DOI: 10.1038/s41467-023-42652-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Identifying oncological applications for drugs that are already approved for other medical indications is considered a possible solution for the increasing costs of cancer treatment. Under the hypothesis that nutritional stress through fasting might enhance the antitumour properties of at least some non-oncological agents, by screening drug libraries, we find that cholesterol biosynthesis inhibitors (CBIs), including simvastatin, have increased activity against cancers of different histology under fasting conditions. We show fasting's ability to increase CBIs' antitumour effects to depend on the reduction in circulating insulin, insulin-like growth factor-1 and leptin, which blunts the expression of enzymes from the cholesterol biosynthesis pathway and enhances cholesterol efflux from cancer cells. Ultimately, low cholesterol levels through combined fasting and CBIs reduce AKT and STAT3 activity, oxidative phosphorylation and energy stores in the tumour. Our results support further studies of CBIs in combination with fasting-based dietary regimens in cancer treatment and highlight the value of fasting for drug repurposing in oncology.
Collapse
Affiliation(s)
- Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Ana Guijarro
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, Via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, 43125, Parma, Italy
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Lizzia Raffaghello
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Roberto Benelli
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Pamela Becherini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Asmaa Namatalla
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
| | - Daniele Reverberi
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
86
|
Wang L, Wang YJ, Wang R, Gong FL, Shi YH, Li SN, Chen PP, Yuan YF. Fasting mimicking diet inhibits tumor-associated macrophage survival and pro-tumor function in hypoxia: implications for combination therapy with anti-angiogenic agent. J Transl Med 2023; 21:754. [PMID: 37884960 PMCID: PMC10601181 DOI: 10.1186/s12967-023-04577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Recent research shows that tumor-associated macrophages (TAMs) are the primary consumers of glucose in tumor tissue, surpassing that of tumor cells. Our previous studies revealed that inhibiting glucose uptake impairs the survival and tumor-promoting function of hypoxic TAMs, suggesting that glucose reduction by energy restriction (calorie restriction or short-term fasting) may has a significant impact on TAMs. The purpose of this study is to verify the effect of fasting-mimicking diet (FMD) on TAMs, and to determine whether FMD synergizes with anti-angiogenic drug apatinib via TAMs. METHODS The effect of FMD on TAMs and its synergistic effects with apatinib were observed using an orthotopic mouse breast cancer model. An in vitro cell model, utilizing M2 macrophages derived from THP-1 cell line, was intended to assess the effects of low glucose on TAMs under hypoxic and normoxic conditions. Bioinformatics was used to screen for potential mechanisms of action, which were then validated both in vivo and in vitro. RESULTS FMD significantly inhibit the pro-tumor function of TAMs in vivo and in vitro, with the inhibitory effect being more pronounced under hypoxic conditions. Additionally, the combination of FMD-mediated TAMs inhibition with apatinib results in synergistic anti-tumor activity. This effect is partially mediated by the downregulation of CCL8 expression and secretion by the mTOR-HIF-1α signaling pathway. CONCLUSIONS These results support further clinical combination studies of FMD and anti-angiogenic therapy as potential anti-tumor strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Yu-Jie Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Rong Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Fu-Lian Gong
- School of Medicine, National Children's Medical Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Shanghai, 200127, China
| | - Yu-Huan Shi
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Sheng-Nan Li
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Pan-Pan Chen
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Yong-Fang Yuan
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China.
| |
Collapse
|
87
|
Udumula MP, Singh H, Rashid F, Poisson L, Tiwari N, Dimitrova I, Hijaz M, Gogoi R, Swenor M, Munkarah A, Giri S, Rattan R. Intermittent fasting induced ketogenesis inhibits mouse epithelial ovarian cancer by promoting antitumor T cell response. iScience 2023; 26:107839. [PMID: 37822507 PMCID: PMC10562806 DOI: 10.1016/j.isci.2023.107839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/28/2023] [Accepted: 09/02/2023] [Indexed: 10/13/2023] Open
Abstract
In various cancer models, dietary interventions have been shown to inhibit tumor growth, improve anticancer drug efficacy, and enhance immunity, but no such evidence exists for epithelial ovarian cancer (EOC), the most lethal gynecologic cancer. The anticancer immune responses induced by 16-h intermittent fasting (IF) were studied in mice with EOC. IF consistently reduced metabolic growth factors and cytokines that stimulate tumor growth, creating a tumor-hostile environment. Immune profiling showed that IF dramatically alters anti-cancer immunity by increasing CD4+ and CD8+ cells, Th1 and cytotoxic responses, and metabolic fitness. β-hydroxy butyrate (BHB), a bioactive metabolite produced by IF, partially imitates its anticancer effects by inducing CD8+ effector function. In a direct comparison, IF outperformed exogenous BHB treatment in survival and anti-tumor immune response, probably due to increased ketogenesis. Thus, IF and one of its metabolic mediators BHB suppress EOC growth and sustain a potent anti-tumor T cell response.
Collapse
Affiliation(s)
- Mary Priyanka Udumula
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Harshit Singh
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Faraz Rashid
- Metabolomics Core, Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Laila Poisson
- Department of Public Health Services and Center for Bioinformatics and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Nivedita Tiwari
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Irina Dimitrova
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Miriana Hijaz
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Radhika Gogoi
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Margaret Swenor
- Department of Lifestyle and Functional Medicine, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Adnan Munkarah
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Shailendra Giri
- Metabolomics Core, Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Ramandeep Rattan
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
- Department of Ob/Gyn, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
88
|
Ivessa AS, Singh S. The increase in cell death rates in caloric restricted cells of the yeast helicase mutant rrm3 is Sir complex dependent. Sci Rep 2023; 13:17832. [PMID: 37857740 PMCID: PMC10587150 DOI: 10.1038/s41598-023-45125-z] [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: 11/29/2022] [Accepted: 10/16/2023] [Indexed: 10/21/2023] Open
Abstract
Calorie restriction (CR), which is a reduction in calorie intake without malnutrition, usually extends lifespan and improves tissue integrity. This report focuses on the relationship between nuclear genomic instability and dietary-restriction and its effect on cell survival. We demonstrate that the cell survival rates of the genomic instability yeast mutant rrm3 change under metabolic restricted conditions. Rrm3 is a DNA helicase, chromosomal replication slows (and potentially stalls) in its absence with increased rates at over 1400 natural pause sites including sites within ribosomal DNA and tRNA genes. Whereas rrm3 mutant cells have lower cell death rates compared to wild type (WT) in growth medium containing normal glucose levels (i.e., 2%), under CR growth conditions cell death rates increase in the rrm3 mutant to levels, which are higher than WT. The silent-information-regulatory (Sir) protein complex and mitochondrial oxidative stress are required for the increase in cell death rates in the rrm3 mutant when cells are transferred from growth medium containing 2% glucose to CR-medium. The Rad53 checkpoint protein is highly phosphorylated in the rrm3 mutant in response to genomic instability in growth medium containing 2% glucose. Under CR, Rad53 phosphorylation is largely reduced in the rrm3 mutant in a Sir-complex dependent manner. Since CR is an adjuvant treatment during chemotherapy, which may target genomic instability in cancer cells, our studies may gain further insight into how these therapy strategies can be improved.
Collapse
Affiliation(s)
- Andreas S Ivessa
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, 185 South Orange Avenue, Newark, NJ, 07101-1709, USA.
| | - Sukhwinder Singh
- Pathology and Laboratory Medicine/Flow Cytometry and Immunology Core Laboratory, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, 185 South Orange Avenue, Newark, NJ, 07101-1709, USA
| |
Collapse
|
89
|
Becker S, Momoh J, Biancacci I, Möckel D, Wang Q, May JN, Su H, Candels LS, Berres ML, Kiessling F, Hatting M, Lammers T, Trautwein C. Intermittent Fasting Primes the Tumor Microenvironment and Improves Nanomedicine Delivery in Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208042. [PMID: 37376850 DOI: 10.1002/smll.202208042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/18/2023] [Indexed: 06/29/2023]
Abstract
Fasting has many health benefits, including reduced chemotherapy toxicity and improved efficacy. It is unclear how fasting affects the tumor microenvironment (TME) and tumor-targeted drug delivery. Here the effects of intermittent (IF) and short-term (STF) fasting are investigated on tumor growth, TME composition, and liposome delivery in allogeneic hepatocellular carcinoma (HCC) mouse models. To this end, mice are inoculated either subcutaneously or intrahepatically with Hep-55.1C cells and subjected to IF for 24 d or to STF for 1 d. IF but not STF significantly slows down tumor growth. IF increases tumor vascularization and decreases collagen density, resulting in improved liposome delivery. In vitro, fasting furthermore promotes the tumor cell uptake of liposomes. These results demonstrate that IF shapes the TME in HCC towards enhanced drug delivery. Finally, when combining IF with liposomal doxorubicin treatment, the antitumor efficacy of nanochemotherapy is found to be increased, while systemic side effects are reduced. Altogether, these findings exemplify that the beneficial effects of fasting on anticancer therapy outcomes go beyond modulating metabolism at the molecular level.
Collapse
Affiliation(s)
- Svea Becker
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Jeffrey Momoh
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Ilaria Biancacci
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Diana Möckel
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Qingbi Wang
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Jan-Niklas May
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Huan Su
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Lena Susanna Candels
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Marie-Luise Berres
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Maximilian Hatting
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Christian Trautwein
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| |
Collapse
|
90
|
Cortellino S, Quagliariello V, Delfanti G, Blaževitš O, Chiodoni C, Maurea N, Di Mauro A, Tatangelo F, Pisati F, Shmahala A, Lazzeri S, Spagnolo V, Visco E, Tripodo C, Casorati G, Dellabona P, Longo VD. Fasting mimicking diet in mice delays cancer growth and reduces immunotherapy-associated cardiovascular and systemic side effects. Nat Commun 2023; 14:5529. [PMID: 37684243 PMCID: PMC10491752 DOI: 10.1038/s41467-023-41066-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Immune checkpoint inhibitors cause side effects ranging from autoimmune endocrine disorders to severe cardiotoxicity. Periodic Fasting mimicking diet (FMD) cycles are emerging as promising enhancers of a wide range of cancer therapies including immunotherapy. Here, either FMD cycles alone or in combination with anti-OX40/anti-PD-L1 are much more effective than immune checkpoint inhibitors alone in delaying melanoma growth in mice. FMD cycles in combination with anti-OX40/anti-PD-L1 also show a trend for increased effects against a lung cancer model. As importantly, the cardiac fibrosis, necrosis and hypertrophy caused by immune checkpoint inhibitors are prevented/reversed by FMD treatment in both cancer models whereas immune infiltration of CD3+ and CD8+ cells in myocardial tissues and systemic and myocardial markers of oxidative stress and inflammation are reduced. These results indicate that FMD cycles in combination with immunotherapy can delay cancer growth while reducing side effects including cardiotoxicity.
Collapse
Affiliation(s)
- S Cortellino
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - V Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - G Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - O Blaževitš
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - C Chiodoni
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - N Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - A Di Mauro
- Pathology and Cytopathology Unit, Department of Support to Cancer Pathways Diagnostics Area, Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale", 80131, Naples, Italy
| | - F Tatangelo
- Pathology and Cytopathology Unit, Department of Support to Cancer Pathways Diagnostics Area, Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale", 80131, Naples, Italy
| | - F Pisati
- Histopathology Unit, Cogentech Società Benefit srl, 20139, Milan, Italy
| | - A Shmahala
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - S Lazzeri
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - V Spagnolo
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - E Visco
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - C Tripodo
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- University of Palermo School of Medicine, Palermo, Italy
| | - G Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - P Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - V D Longo
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
91
|
Huang W, Li X, Song H, Yin Y, Wang H. Verification of fasting-mimicking diet to assist monotherapy of human cancer-bearing models. Biochem Pharmacol 2023; 215:115699. [PMID: 37482198 DOI: 10.1016/j.bcp.2023.115699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
The efficacy of a single clinical nanodrug for cancer treatment is still unsatisfactory, especially for drug-resistant cancer. Herein, we applied a fasting-mimicking diet (FMD) approach via dietary intervention to assist single clinical nanodrug for breast or ovarian cancer treatments instead of using multi-drug therapies which might cause adverse side effects. Specifically, we adopted Doxil or Abraxane to treat human breast tumor-bearing nude mice and Doxil to treat the human ovarian tumor and drug-resistant ovarian tumor-bearing nude mice under FMD conditions, respectively. According to the results, the FMD condition can promote the cellular uptake and cytotoxicity of a single nanodrug, reduce the ATP level in drug-resistant tumor cells to hinder drug efflux, normalize tumor blood vessels, relieve tumor hypoxia, and increase the accumulation of nanodrugs at tumor sites, thereby enhancing the therapeutic effects on these types of human cancers. Collectively, these results demonstrate that the FMD strategy of significance can become a practical, alternative, and promising assistant for single nanodrug for enhancing cancer therapy and clinical translation.
Collapse
Affiliation(s)
- Wenping Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyang Li
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Haohao Song
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yue Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
92
|
Xiao Y, Yu TJ, Xu Y, Ding R, Wang YP, Jiang YZ, Shao ZM. Emerging therapies in cancer metabolism. Cell Metab 2023; 35:1283-1303. [PMID: 37557070 DOI: 10.1016/j.cmet.2023.07.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/20/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023]
Abstract
Metabolic reprogramming in cancer is not only a biological hallmark but also reveals treatment vulnerabilities. Numerous metabolic molecules have shown promise as treatment targets to impede tumor progression in preclinical studies, with some advancing to clinical trials. However, the intricacy and adaptability of metabolic networks hinder the effectiveness of metabolic therapies. This review summarizes the metabolic targets for cancer treatment and provides an overview of the current status of clinical trials targeting cancer metabolism. Additionally, we decipher crucial factors that limit the efficacy of metabolism-based therapies and propose future directions. With advances in integrating multi-omics, single-cell, and spatial technologies, as well as the ability to track metabolic adaptation more precisely and dynamically, clinicians can personalize metabolic therapies for improved cancer treatment.
Collapse
Affiliation(s)
- Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tian-Jian Yu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Rui Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Ping Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
93
|
Li J, Fu R, Guo X, Pan Z, Xie J. Acupuncture improves immunity and fatigue after chemotherapy in breast cancer patients by inhibiting the Leptin/AMPK signaling pathway. Support Care Cancer 2023; 31:506. [PMID: 37542585 PMCID: PMC10404187 DOI: 10.1007/s00520-023-07967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/24/2023] [Indexed: 08/07/2023]
Abstract
OBJECTIVE Acupuncture has become a popular complementary treatment in oncology. This study is based on RNA-Seq transcriptome sequencing technology to investigate the molecular mechanisms underlying the effect of acupuncture-mediated regulation of the Leptin/AMPK signaling pathway on mitochondrial dysfunction-induced fatigue in breast cancer patients after chemotherapy. METHODS Peripheral blood samples from 10 patients with post-operative chemotherapy for breast cancer were selected for transcriptome sequencing to screen the key molecular pathways involved in fatigue after chemotherapy in breast cancer patients. Besides, peripheral blood samples were collected from 138 post-operative chemotherapy patients with breast cancer to study the composite fatigue and quality of life scores. Flow cytometry was used to detect T lymphocyte subsets in peripheral blood-specific immune cells. In addition, a blood cell analyzer was used to measure peripheral blood leukocyte counts, and MSP-PCR was used to detect mitochondrial DNA mutations in peripheral blood leukocytes. RESULTS Transcriptome bioinformatics analysis screened 147 up-regulated mRNAs and 160 down-regulated mRNAs. Leptin protein was confirmed as the key factor. Leptin was significantly higher in the peripheral blood of breast cancer patients who developed fatigue after chemotherapy. Acupuncture treatment effectively improved post-chemotherapy fatigue and immune status in breast cancer patients, suppressed the expression of Leptin/AMPK signaling pathway-related factor and leukocyte counts, and significantly reduced the rate of mitochondrial DNA mutations in peripheral blood leukocytes. CONCLUSION The Leptin/AMPK signaling pathway may be the key molecular pathway affecting the occurrence of fatigue after chemotherapy in breast cancer patients. Leptin may improve post-chemotherapy fatigue in breast cancer patients by activating AMPK phosphorylation and alleviating mitochondrial functional impairment.
Collapse
Affiliation(s)
- Jinxia Li
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Ruiyang Fu
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Xiaoqing Guo
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Zhongqiang Pan
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Jingjun Xie
- Department of Rehabilitation Medicine, The First People's Hospital of Huzhou, No. 158, Guangchang Hou Road, Huzhou, 313000, Zhejiang, People's Republic of China.
| |
Collapse
|
94
|
Hanslian E, Koppold D, Michalsen A. [Fasting - a potent modern therapy]. Dtsch Med Wochenschr 2023; 148:1043-1053. [PMID: 37541295 DOI: 10.1055/a-2119-3516] [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: 08/06/2023]
Abstract
Therapeutic fasting has found its way into modern medicine in the last decade through a multitude of experimental work and animal studies as well as increasing clinical research. It is a procedure with a tradition dating back thousands of years and thus comes with a variety of different practices. What they all have in common, is the reduction of daily food intake for a limited period of time. This has a variety of effects on metabolism, cells and organ systems, which can make it a potent tool in medical practice.
Collapse
Affiliation(s)
- Etienne Hanslian
- Institut für Sozialmedizin, Epidemiologie und Gesundheitsökonomie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Andreas Michalsen
- Institut für Sozialmedizin, Epidemiologie und Gesundheitsökonomie, Charité Universitätsmedizin Berlin, Berlin, Germany
- Naturheilkunde, Immanuel Krankenhaus Berlin Standort Berlin-Wannsee, Berlin, Germany
| |
Collapse
|
95
|
Cortellino S, Longo VD. Metabolites and Immune Response in Tumor Microenvironments. Cancers (Basel) 2023; 15:3898. [PMID: 37568713 PMCID: PMC10417674 DOI: 10.3390/cancers15153898] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The remodeled cancer cell metabolism affects the tumor microenvironment and promotes an immunosuppressive state by changing the levels of macro- and micronutrients and by releasing hormones and cytokines that recruit immunosuppressive immune cells. Novel dietary interventions such as amino acid restriction and periodic fasting mimicking diets can prevent or dampen the formation of an immunosuppressive microenvironment by acting systemically on the release of hormones and growth factors, inhibiting the release of proinflammatory cytokines, and remodeling the tumor vasculature and extracellular matrix. Here, we discuss the latest research on the effects of these therapeutic interventions on immunometabolism and tumor immune response and future scenarios pertaining to how dietary interventions could contribute to cancer therapy.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Valter D. Longo
- IFOM, The AIRC Institute of Molecular Oncology, 20139 Milan, Italy
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
96
|
Akinsulie OC, Shahzad S, Ogunleye SC, Oladapo IP, Joshi M, Ugwu CE, Gbadegoye JO, Hassan FO, Adeleke R, Afolabi Akande Q, Adesola RO. Crosstalk between hypoxic cellular micro-environment and the immune system: a potential therapeutic target for infectious diseases. Front Immunol 2023; 14:1224102. [PMID: 37600803 PMCID: PMC10434535 DOI: 10.3389/fimmu.2023.1224102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
There are overwhelming reports on the promotional effect of hypoxia on the malignant behavior of various forms of cancer cells. This has been proposed and tested exhaustively in the light of cancer immunotherapy. However, there could be more interesting functions of a hypoxic cellular micro-environment than malignancy. There is a highly intricate crosstalk between hypoxia inducible factor (HIF), a transcriptional factor produced during hypoxia, and nuclear factor kappa B (NF-κB) which has been well characterized in various immune cell types. This important crosstalk shares common activating and inhibitory stimuli, regulators, and molecular targets. Impaired hydroxylase activity contributes to the activation of HIFs. Inflammatory ligands activate NF-κB activity, which leads to the expression of inflammatory and anti-apoptotic genes. The eventual sequelae of the interaction between these two molecular players in immune cells, either bolstering or abrogating functions, is largely cell-type dependent. Importantly, this holds promise for interesting therapeutic interventions against several infectious diseases, as some HIF agonists have helped prevent immune-related diseases. Hypoxia and inflammation are common features of infectious diseases. Here, we highlighted the role of this crosstalk in the light of functional immunity against infection and inflammation, with special focus on various innate and adaptive immune cells. Particularly, we discussed the bidirectional effects of this crosstalk in the regulation of immune responses by monocytes/macrophages, dendritic cells, neutrophils, B cells, and T cells. We believe an advanced understanding of the interplay between HIFs and NF-kB could reveal novel therapeutic targets for various infectious diseases with limited treatment options.
Collapse
Affiliation(s)
- Olalekan Chris Akinsulie
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sammuel Shahzad
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Seto Charles Ogunleye
- College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Ifeoluwa Peace Oladapo
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Melina Joshi
- Center for Molecular Dynamics Nepal, Kathmandu, Nepal
| | - Charles Egede Ugwu
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
| | - Joy Olaoluwa Gbadegoye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fasilat Oluwakemi Hassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Richard Adeleke
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Qudus Afolabi Akande
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | | |
Collapse
|
97
|
Rawat V, DeLear P, Prashanth P, Ozgurses ME, Tebeje A, Burns PA, Conger KO, Solís C, Hasnain Y, Novikova A, Endress JE, González-Sánchez P, Dong W, Stephanopoulos G, DeNicola GM, Harris IS, Sept D, Mason FM, Coloff JL. Drug screening in human physiologic medium identifies uric acid as an inhibitor of rigosertib efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550731. [PMID: 37546939 PMCID: PMC10402161 DOI: 10.1101/2023.07.26.550731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The non-physiological nutrient levels found in traditional culture media have been shown to affect numerous aspects of cancer cell physiology, including how cells respond to certain therapeutic agents. Here, we comprehensively evaluated how physiological nutrient levels impact therapeutic response by performing drug screening in human plasma-like medium (HPLM). We observed dramatic nutrient-dependent changes in sensitivity to a variety of FDA-approved and clinically trialed compounds, including rigosertib, an experimental cancer therapeutic that has recently failed in phase 3 clinical trials. Mechanistically, we found that the ability of rigosertib to destabilize microtubules is strongly inhibited by the purine metabolism waste product uric acid, which is uniquely abundant in humans relative to traditional in vitro and in vivo cancer models. Structural modelling studies suggest that uric acid interacts with the tubulin-rigosertib complex and may act as an uncompetitive inhibitor of rigosertib. These results offer a possible explanation for the failure of rigosertib in clinical trials and demonstrate the utility of physiological media to achieve in vitro results that better represent human therapeutic responses.
Collapse
Affiliation(s)
- Vipin Rawat
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | - Patrick DeLear
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Prarthana Prashanth
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | - Mete Emir Ozgurses
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | - Anteneh Tebeje
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Philippa A. Burns
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | - Kelly O. Conger
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | - Christopher Solís
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL
| | - Yasir Hasnain
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | - Anna Novikova
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| | | | | | - Wentao Dong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Greg Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL
| | - Isaac S. Harris
- Department of Biomedical Genetics, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - David Sept
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Frank M. Mason
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan L. Coloff
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL
| |
Collapse
|
98
|
Chetta P, Sriram R, Zadra G. Lactate as Key Metabolite in Prostate Cancer Progression: What Are the Clinical Implications? Cancers (Basel) 2023; 15:3473. [PMID: 37444583 PMCID: PMC10340474 DOI: 10.3390/cancers15133473] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Advanced prostate cancer represents the fifth leading cause of cancer death in men worldwide. Although androgen-receptor signaling is the major driver of the disease, evidence is accumulating that disease progression is supported by substantial metabolic changes. Alterations in de novo lipogenesis and fatty acid catabolism are consistently reported during prostate cancer development and progression in association with androgen-receptor signaling. Therefore, the term "lipogenic phenotype" is frequently used to describe the complex metabolic rewiring that occurs in prostate cancer. However, a new scenario has emerged in which lactate may play a major role. Alterations in oncogenes/tumor suppressors, androgen signaling, hypoxic conditions, and cells in the tumor microenvironment can promote aerobic glycolysis in prostate cancer cells and the release of lactate in the tumor microenvironment, favoring immune evasion and metastasis. As prostate cancer is composed of metabolically heterogenous cells, glycolytic prostate cancer cells or cancer-associated fibroblasts can also secrete lactate and create "symbiotic" interactions with oxidative prostate cancer cells via lactate shuttling to sustain disease progression. Here, we discuss the multifaceted role of lactate in prostate cancer progression, taking into account the influence of the systemic metabolic and gut microbiota. We call special attention to the clinical opportunities of imaging lactate accumulation for patient stratification and targeting lactate metabolism.
Collapse
Affiliation(s)
- Paolo Chetta
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Giorgia Zadra
- Institute of Molecular Genetics, National Research Council (IGM-CNR), 27100 Pavia, Italy
| |
Collapse
|
99
|
Maragkakis M, Malla S, Hatzoglou M, Trifunovic A, Glick AB, Finkel T, Longo VD, Kaushik S, Muñoz-Cánoves P, Lithgow GJ, Naidoo N, Booth LN, Payea MJ, Herman AB, de Cabo R, Wilson DM, Ferrucci L, Gorospe M. Biology of Stress Responses in Aging. AGING BIOLOGY 2023; 1:20230002. [PMID: 38500537 PMCID: PMC10947073 DOI: 10.59368/agingbio.20230001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
On April 28th, 2022, a group of scientific leaders gathered virtually to discuss molecular and cellular mechanisms of responses to stress. Conditions of acute, high-intensity stress are well documented to induce a series of adaptive responses that aim to promote survival until the stress has dissipated and then guide recovery. However, high-intensity or persistent stress that goes beyond the cell's compensatory capacity are countered with resilience strategies that are not completely understood. These adaptative strategies, which are an essential component of the study of aging biology, were the theme of the meeting. Specific topics discussed included mechanisms of proteostasis, such as the unfolded protein response (UPR) and the integrated stress response (ISR), as well as mitochondrial stress and lysosomal stress responses. Attention was also given to regulatory mechanisms and associated biological processes linked to age-related conditions, such as muscle loss and regeneration, cancer, senescence, sleep quality, and degenerative disease, with a general focus on the relevance of stress responses to frailty. We summarize the concepts and potential future directions that emerged from the discussion and highlight their relevance to the study of aging and age-related chronic diseases.
Collapse
Affiliation(s)
- Manolis Maragkakis
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Sulochan Malla
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Maria Hatzoglou
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Aleksandra Trifunovic
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Adam B Glick
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Toren Finkel
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Valter D Longo
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Susmita Kaushik
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Pura Muñoz-Cánoves
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Gordon J Lithgow
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Nirinjini Naidoo
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Lauren N Booth
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Matthew J Payea
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Allison B Herman
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Rafael de Cabo
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - David M Wilson
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Luigi Ferrucci
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Myriam Gorospe
- National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
100
|
Zheng P, Lin Z, Ding Y, Duan S. Targeting the dynamics of cancer metabolism in the era of precision oncology. Metabolism 2023:155615. [PMID: 37286129 DOI: 10.1016/j.metabol.2023.155615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Cancer metabolic reprogramming is a promising target for cancer therapy. The progression of tumors, including their growth, development, metastasis, and spread, is a dynamic process that varies over time and location. This means that the metabolic state of tumors also fluctuates. A recent study found that energy production efficiency is lower in solid tumors but increases significantly in tumor metastasis. Despite its importance for targeted tumor metabolism therapy, few studies have described the dynamic metabolic changes of tumors. In this commentary, we discuss the limitations of past targeted tumor metabolism therapy and the key findings of this study. We also summarize its immediate clinical implications for dietary intervention and explore future research directions for understanding the dynamic changes in tumor metabolic reprogramming.
Collapse
Affiliation(s)
- Peijie Zheng
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
| | - Zihao Lin
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
| | - Yuemin Ding
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, China; Institute of Translational Medicine, Hangzhou City University, Hangzhou 310015, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, China.
| | - Shiwei Duan
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, China; Institute of Translational Medicine, Hangzhou City University, Hangzhou 310015, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, China.
| |
Collapse
|