1
|
Ben Kridis W, Hadj Taieb E, Ben Lassoued R, Khanfir A. Fasting during cancer chemotherapy: single centre study. BMJ Support Palliat Care 2025:spcare-2025-005369. [PMID: 40175058 DOI: 10.1136/spcare-2025-005369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Affiliation(s)
| | | | | | - Afef Khanfir
- Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia
| |
Collapse
|
2
|
Fatima G, Mehdi AA, Fedacko J, Hadi N, Magomedova A, Mehdi A. Fasting as Cancer Treatment: Myth or Breakthrough in Oncology. Cureus 2025; 17:e81395. [PMID: 40296920 PMCID: PMC12035504 DOI: 10.7759/cureus.81395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The concept of fasting as a potential cancer treatment has garnered increasing interest, particularly in light of emerging evidence linking dietary interventions to cancer progression and therapy outcomes. This article explores whether fasting, either intermittent or prolonged, can be a viable standalone treatment for cancer or if its therapeutic potential lies in its adjunctive role. Current research suggests that fasting induces a metabolic shift, which may inhibit cancer cell proliferation by depriving them of essential nutrients. Additionally, fasting has been shown to enhance the body's stress resistance, promote autophagy, and possibly make cancer cells more vulnerable to standard treatments such as chemotherapy and radiotherapy. However, the application of fasting as a sole treatment for cancer remains controversial and lacks substantial clinical validation. While animal models and in vitro studies indicate promising results, the translation to human trials is complex, with various types of cancer responding differently to dietary interventions. Moreover, concerns about malnutrition, loss of muscle mass, and the overall health of cancer patients undergoing fasting without supervision must be addressed. The paper critically examines the myth and reality surrounding fasting as a cancer treatment, reviewing key studies and clinical trials to provide a comprehensive understanding of its efficacy and safety. While fasting may hold promise as a supportive therapy, particularly in combination with traditional treatments, there is currently insufficient evidence to support its use as a primary treatment modality. Further research is needed to establish the parameters in which fasting might be beneficial, such as specific cancer types, patient populations, and optimal fasting regimens. Thus, while the idea of fasting as a cancer breakthrough is compelling, it remains a complementary approach rather than a standalone solution in oncology.
Collapse
Affiliation(s)
- Ghizal Fatima
- Department of Public Health, Era's Lucknow Medical College and Hospital, Era University, Lucknow, IND
| | - Abbas A Mehdi
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, IND
| | - Jan Fedacko
- Department of Cardiology, Pavol Jozef Safarik University, Kosice, Kosice, SVK
| | - Najah Hadi
- Department of Medicine, University of Kufa, Najaf, IRQ
| | | | - Ammar Mehdi
- Department of Pediatric Dentistry, Career Post Graduate Institute of Dental Sciences and Hospital, Lucknow, IND
| |
Collapse
|
3
|
Lu P, Tsang T, Badowski MS, Pennington ME, Meade‐Tollin LC. Evaluation of the Clinical Safety of the Low-Cost Warburg Therapy for the Treatment of Patients With Advanced Cancers. Cancer Med 2024; 13:e70469. [PMID: 39629677 PMCID: PMC11615646 DOI: 10.1002/cam4.70469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Rising cancer care costs are becoming cost prohibitive for lower income people worldwide. We developed the Warburg protocol as a low-cost option for the treatment of cancer that was inspired. It was developed to exploit an Achilles heel which is a hallmark of cancer cells; the metabolic requirement for higher levels of glucose than normal cells. OBJECTIVE The purpose of this report is to assess the clinical safety and affordability of the Warburg therapy as an option for patients with advanced cancers. METHODS Between 2021 and 2023, 251 patients with advanced cancers received a total of 8542 treatments with the Warburg therapy. To restrict the supply of blood glucose to cancerous tumors, regular human insulin was administered (IV) sufficient to reduce blood glucose concentrations to hypoglycemic levels for 40-60 min. Subroutine doses of fluorouracil and cyclophosphamide were administered intravenously during this hypoglycemic period. Food or intravenous glucose was given as needed to return blood glucose to euglycemic levels after treatment. Patient symptoms, status, vitals, blood glucose, and hypoglycemic symptoms were monitored throughout treatment. Various blood parameters were measured before and after patients' course of treatment. RESULTS There were no irreversible adverse reactions in advanced tumor patients of different ages and different cancer types after treatment. There was no significant fluctuation in blood glucose levels in diabetic and non-diabetic patients after treatment, and the weight, vital index and blood biochemical index of patients before and after multiple treatments exhibited little variation. CONCLUSION Warburg therapy for the treatment of advanced tumors is clinically feasible, and safe for multiple treatments. It is inexpensive and widely applicable to different patient groups.
Collapse
Affiliation(s)
- Peihua Lu
- Department of Hematology and OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiChina
- School of MedicineJiangnan UniversityWuxiChina
| | - Tom Tsang
- American Goodwill Mission to China Inc. 501(c) (3)TucsonArizonaUSA
- Warburg MedicalChongqingChina
| | | | | | - Linda C. Meade‐Tollin
- American Goodwill Mission to China Inc. 501(c) (3)TucsonArizonaUSA
- Department of SurgeryUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
4
|
Dan X, He Y, Tian Y, Chen T, Yu J. Summary of Evidence on Nutritional Management for Patients Undergoing Chemotherapy. Cancer Med 2024; 13:e70519. [PMID: 39698953 PMCID: PMC11656406 DOI: 10.1002/cam4.70519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE This paper aims to consolidate the most robust evidence on nutritional strategies for patients undergoing chemotherapy, offering evidence-based guidance for clinical practice. The review highlights critical evidence gaps in nutritional therapy for advanced gastric cancer (AGC) patients undergoing systemic therapy, integrating findings from both prospective and retrospective studies. METHOD According to the "6S" evidence resource pyramid model, clinical decision-making tools, guidelines, expert consensus, and systematic reviews on nutritional management for chemotherapy patients were systematically retrieved from national and international databases. The methodological quality of the selected literature was evaluated using AGREE II for guidelines, the JBI Evidence-Based Healthcare Center's standards for systematic reviews, and expert consensus developed by evidence-based practice experts. RESULTS A total of 47 articles were analyzed, consisting of 12 guidelines, 12 expert consensus statements, and 23 systematic reviews. The findings were categorized into five dimensions: interdisciplinary collaboration, nutritional screening and assessment, nutritional requirements, nutritional therapy, and discharge and follow-up, resulting in the identification of 62 pieces of relevant evidence. CONCLUSIONS The study provides comprehensive, evidence-based recommendations for nutritional management in chemotherapy patients. Application of the evidence should be adapted to specific clinical scenarios, patient conditions, preferences, and expert judgment to ensure both feasibility and relevance in clinical practice. CONTRIBUTIONS This review consolidates diverse nutritional management strategies into a unified framework, addressing evidence gaps in AGC under systemic therapy. Integrating prospective and retrospective studies with interdisciplinary insights provides evidence-based recommendations to enhance patient care through personalized and standardized approaches.
Collapse
Affiliation(s)
- Xin Dan
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University HospitalSichuan UniversityChengduSichuanChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduSichuanChina
| | - Ya‐Lin He
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University HospitalSichuan UniversityChengduSichuanChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduSichuanChina
| | - Ya‐Lin Tian
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University HospitalSichuan UniversityChengduSichuanChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduSichuanChina
| | - Tang‐Lin Chen
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University HospitalSichuan UniversityChengduSichuanChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduSichuanChina
| | - Jia‐Yi Yu
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University HospitalSichuan UniversityChengduSichuanChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduSichuanChina
| |
Collapse
|
5
|
Meng Y, Sun J, Zhang G, Yu T, Piao H. Fasting: A Complex, Double-Edged Blade in the Battle Against Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024; 24:1395-1409. [PMID: 39354217 DOI: 10.1007/s12012-024-09925-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
In recent years, there has been a surge in the popularity of fasting as a method to enhance one's health and overall well-being. Fasting is a customary practice characterized by voluntary refraining from consuming food and beverages for a specified duration, ranging from a few hours to several days. The potential advantages of fasting, including enhanced insulin sensitivity, decreased inflammation, and better cellular repair mechanisms, have been well documented. However, the effects of fasting on cancer therapy have been the focus of recent scholarly investigations. Doxorubicin (Dox) is one of the most widely used chemotherapy medications for cancer treatment. Unfortunately, cardiotoxicity, which may lead to heart failure and other cardiovascular issues, has been linked to Dox usage. This study aims to comprehensively examine the possible advantages and disadvantages of fasting concerning Dox-induced cardiotoxicity. Researchers have investigated the potential benefits of fasting in lowering the risk of Dox-induced cardiac damage to solve this problem. Nevertheless, new studies indicate that prolonged alternate-day fasting may adversely affect the heart's capacity to manage the cardiotoxic properties of Dox. Though fasting may benefit overall health, it is essential to proceed cautiously and consider the potential risks in certain circumstances.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Tao Yu
- Department of Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Province Cancer Hospital, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Liaoning Province Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
6
|
He C, Wang Z, Yu J, Mao S, Xiang X. Current Drug Resistance Mechanisms and Treatment Options in Gastrointestinal Stromal Tumors: Summary and Update. Curr Treat Options Oncol 2024; 25:1390-1405. [PMID: 39441520 PMCID: PMC11541409 DOI: 10.1007/s11864-024-01272-7] [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] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
OPINION STATEMENT Gastrointestinal stromal tumor (GIST) is characterized by well-defined oncogenes. Despite the significant improvement in treatment outcomes with adjuvant imatinib therapy for patients, drug resistance remains a major challenge for GIST therapy. This review focuses on the mechanisms contributing to drug resistance phenotype in GIST, such as primary imatinib-resistant mutants, secondary mutations, non-covalent binding of TKI to its target, tumor heterogeneity, re-activation of pro-survival/proliferation pathways through non-KIT/PDGFRA kinases, and loss of therapeutic targets in wild-type GIST. Corresponding suggestions are proposed to overcome drug-resistance phenotype of GIST. This review also summarizes the suitability of currently approved TKIs on different KIT/PDGFRA mutations and updates related clinical trials. Recent potent drugs and emerging strategies against advanced GISTs in clinical trials are presented. Additionally, metabolic intervention offers a new avenue for clinical management in GIST. A landscape of metabolism in GIST and metabolic changes under imatinib treatment are summarized based on currently published data. The OXPHOS pathway is a promising therapeutic target in combination with TKI against sensitive KIT/PDGFRA mutants. Comprehensive understanding of the above resistance mechanisms, experimental drugs/strategies and metabolic changes is critical to implement the proper therapy strategy and improve the clinical therapy outcomes for GIST.
Collapse
Affiliation(s)
- Chunxiao He
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Zilong Wang
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiaying Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shuang Mao
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xi Xiang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Sheykhbahaei N, Tameemi AHA, Koopaie M. Effect of short-term fasting on the cisplatin activity in human oral squamous cell carcinoma cell line HN5 and chemotherapy side effects. BMC Cancer 2024; 24:989. [PMID: 39123141 PMCID: PMC11316436 DOI: 10.1186/s12885-024-12752-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Ketogenic interventions like short-term fasting show potential as complementary therapies to enhance the effectiveness of chemotherapy for cancer. However, the specific effects of fasting on head and neck squamous cell carcinoma (HNSCC) cells and healthy oral mucosa cells during these treatments are not well understood. This study investigates whether short-term fasting can differentially impact HNSCC cell survival and viability compared to healthy keratinocytes while undergoing standard chemotherapy regimens. METHODS This study investigated the effects of fasting on cell viability in HN5 cell line and healthy oral keratinocyte cells. The HN5 cell line, derived from human tongue squamous cell carcinoma, and primary human keratinocytes isolated from the basal layer of gingival epithelium were divided into three groups: (1) control, (2) treated with the standard chemotherapeutic agent cisplatin, and (3) treated with cisplatin under fasting conditions achieved through 48-hour glucose restriction mimicking the blood glucose levels of fasted individuals. Cell proliferation was assessed at 48 and 72 h using the MTT assay, a colorimetric method based on mitochondrial dehydrogenase activity. Flow cytometry analysis with specific apoptosis and necrosis markers distinguished between early and late apoptotic, necrotic, and viable cells. RESULTS Cell viability in HN5 and healthy keratinocyte cells decreased in cisplatin with low glucose groups compared to cisplatin and control groups. The same results were observed for healthy keratinocyte cells; only a decrease in cell viability in cisplatin groups compared to control groups was observed, which was not statistically significant. Cell apoptosis in HN5 and healthy keratinocyte cells increased in cisplatin with low glucose groups compared to cisplatin and control groups. In healthy keratinocyte cells, the cisplatin with low glucose group showed an impressive increase in necrosis, late apoptosis, and early apoptosis and a significant decrease in live cells compared with other groups. CONCLUSION This study revealed that short-term fasting chemotherapy significantly improved HNSCC cell line apoptosis and necrosis.
Collapse
Affiliation(s)
- Nafiseh Sheykhbahaei
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, North Kargar St, Tehran, 14399-55991, Iran
| | - Ahmed Hayder Al Tameemi
- Dentist, Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Koopaie
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, North Kargar St, Tehran, 14399-55991, Iran.
| |
Collapse
|
8
|
Tao Y, Gong Z, Shen S, Ding Y, Zan R, Zheng B, Sun W, Ma C, Shu M, Lu X, Liu H, Ni X, Liu H, Suo T. Fasting-induced RNF152 resensitizes gallbladder cancer cells to gemcitabine by inhibiting mTORC1-mediated glycolysis. iScience 2024; 27:109659. [PMID: 38706841 PMCID: PMC11068552 DOI: 10.1016/j.isci.2024.109659] [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: 10/17/2023] [Revised: 01/05/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
Abnormal mTORC1 activation by the lysosomal Ragulator complex has been implicated in cancer and glycolytic metabolism associated with drug resistance. Fasting upregulates RNF152 and mediates the metabolic status of cells. We report that RNF152 regulates mTORC1 signaling by targeting a Ragulator subunit, p18, and attenuates gemcitabine resistance in gallbladder cancer (GBC). We detected levels of RNF152 and p18 in tissues and undertook mechanistic studies using activators, inhibitors, and lentivirus transfections. RNF152 levels were significantly lower in GBC than in adjacent non-cancer tissues. Fasting impairs glycolysis, induces gemcitabine sensitivity, and upregulates RNF152 expression. RNF152 overexpression increases the sensitivity of GBC cells to gemcitabine, whereas silencing RNF152 has the opposite effect. Fasting-induced RNF152 ubiquitinates p18, resulting in proteasomal degradation. RNF152 deficiency increases the lysosomal localization of p18 and increases mTORC1 activity, to promote glycolysis and decrease gemcitabine sensitivity. RNF152 suppresses mTORC1 activity to inhibit glycolysis and enhance gemcitabine sensitivity in GBC.
Collapse
Affiliation(s)
- Ying Tao
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Zijun Gong
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sheng Shen
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yaqi Ding
- Ruijin Hospital LuWan Branch, Shanghai Jiao Tong University School of Medicine Central Laboratory, Shanghai, China
| | - Rui Zan
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bohao Zheng
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wentao Sun
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chaolin Ma
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengxuan Shu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Lu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Han Liu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoling Ni
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Houbao Liu
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Suo
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai, China
- Biliary Tract Disease Institute, Fudan University, Shanghai, China
- The Center of Biliary Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 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
|
10
|
Zhang S, Lv Y, Qian J, Wei W, Zhu Y, Liu Y, Li L, Zhao C, Gao X, Yang Y, Dong J, Gu Y, Chen Y, Sun Q, Jiao X, Lu J, Yan Z, Wang L, Yuan N, Fang Y, Wang J. Adaptive metabolic response to short-term intensive fasting. Clin Nutr 2024; 43:453-467. [PMID: 38181523 DOI: 10.1016/j.clnu.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/19/2023] [Accepted: 12/27/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND & AIMS Short-term intensive fasting (STIF), known as beego in Chinese phonetic articulation, has been practiced for more than two thousand years. However, the potential risk of STIF and the body's response to the risk have not been adequately evaluated. This study aims to address this issue, focusing on the STIF-triggered metabolic response of the liver and kidney. METHODS The STIF procedure in the clinical trial includes a 7-day water-only intensive fasting phase and a 7-day gradual refeeding phase followed by a regular diet. The intensive fasting in humans was assisted with psychological induction. To gain insights not available in the clinical trial, we designed a STIF program for mice that resulted in similar phenotypes seen in humans. Plasma metabolic profiling and examination of gene expression as well as liver and kidney function were performed by omics, molecular, biochemical and flow cytometric analyses. A human cell line model was also used for mechanistic study. RESULTS Clinically significant metabolites of fat and protein were found to accumulate during the fasting phase, but they were relieved after gradual refeeding. Metabolomics profiling revealed a universal pattern in the consumption of metabolic intermediates, in which pyruvate and succinate are the two key metabolites during STIF. In the STIF mouse model, the accumulation of metabolites was mostly counteracted by the upregulation of catabolic enzymes in the liver, which was validated in a human cell model. Kidney filtration function was partially affected by STIF but could be recovered by refeeding. STIF also reduced oxidative and inflammatory levels in the liver and kidney. Moreover, STIF improved lipid metabolism in mice with fatty liver without causing accumulation of metabolites after STIF. CONCLUSIONS The accumulation of metabolites induced by STIF can be relieved by spontaneous upregulation of catabolic enzymes, suggesting an adaptive and protective metabolic response to STIF stress in the mammalian body.
Collapse
Affiliation(s)
- Suping Zhang
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Suzhou Center for Disease Control and Prevention, Suzhou 215004, China
| | - Yaqi Lv
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Jiawei Qian
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Wen Wei
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Susky Life SciTech (Suzhou) Inc., Suzhou 215101, China
| | - Yanfei Zhu
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Yuqing Liu
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Lei Li
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Susky Life SciTech (Suzhou) Inc., Suzhou 215101, China
| | - Chen Zhao
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Susky Life SciTech (Suzhou) Inc., Suzhou 215101, China
| | - Xueqin Gao
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Yanjun Yang
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China
| | - Jin Dong
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Yue Gu
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Yuwei Chen
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China
| | - Qiyuan Sun
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China
| | - Xuehua Jiao
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China
| | - Jie Lu
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China
| | - Zhanjun Yan
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China
| | - Li Wang
- Department of Community Nursing, School of Nursing, Soochow University, Suzhou 215006, China
| | - Na Yuan
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Susky Life SciTech (Suzhou) Inc., Suzhou 215101, China.
| | - Yixuan Fang
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Susky Life SciTech (Suzhou) Inc., Suzhou 215101, China.
| | - Jianrong Wang
- Research Center for Blood Engineering and Manufacturing, Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Susky Life SciTech (Suzhou) Inc., Suzhou 215101, China; Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou 215200, China.
| |
Collapse
|
11
|
David A, Lev-Ari S. Targeting the Gut Microbiome to Improve Immunotherapy Outcomes: A Review. Integr Cancer Ther 2024; 23:15347354241269870. [PMID: 39223798 PMCID: PMC11369881 DOI: 10.1177/15347354241269870] [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] [Revised: 03/18/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
The following narrative review embarks on a comprehensive exploration of the role played by the gut microbiome within the Diet-Microbiota-Immunity (DMI) tripartite, aiming to enhance anti-cancer immunotherapy efficacy. While revolutionizing cancer treatment, resistance to immunotherapy and immune-related adverse events (irAEs) remain challenges. The tumor microenvironment (TME), shaped by cancer cells, influences immunotherapy resistance. The gut microbiome, influenced by genetics, environment, diet, and interventions, emerges as a critical player in TME reshaping, thereby modulating immune responses and treatment outcomes. Dietary patterns like the Mediterranean diet, caloric restriction modifications, and specific nutritional components show promise in influencing the tumor microenvironment and gut microbiome for better treatment outcomes. Antibiotics, disrupting gut microbiota diversity, may compromise immunotherapy efficacy. This review emphasizes the need for tailored nutritional strategies to manipulate microbial communities, enhance immune regulation, and improve immunotherapy accessibility while minimizing side effects. Ongoing studies investigate the impact of dietary interventions on cancer immunotherapy, pointing toward promising developments in personalized cancer care. This narrative review synthesizes existing knowledge and charts a course for future investigations, presenting a holistic perspective on the dynamic interplay between dietary interventions, the gut microbiome, and cancer immunotherapy within the DMI tripartite.
Collapse
Affiliation(s)
- Adi David
- Tal Center for Integrative Medicine, Institute of Oncology, Sheba Medical Center, Ramat-Gan, Israel
| | - Shaked Lev-Ari
- Ella Lemelbaum Institute For Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
- Education Authority, Sheba Medical Center, Ramat-Gan, Israel
| |
Collapse
|
12
|
Shi D, Fang G, Chen Q, Li J, Ruan X, Lian X. Six-hour time-restricted feeding inhibits lung cancer progression and reshapes circadian metabolism. BMC Med 2023; 21:417. [PMID: 37924048 PMCID: PMC10625271 DOI: 10.1186/s12916-023-03131-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Accumulating evidence has suggested an oncogenic effect of diurnal disruption on cancer progression. To test whether targeting circadian rhythm by dietary strategy suppressed lung cancer progression, we adopted 6-h time-restricted feeding (TRF) paradigm to elucidate whether and how TRF impacts lung cancer progression. METHODS This study used multiple lung cancer cell lines, two xenograft mouse models, and a chemical-treated mouse lung cancer model. Stable TIM-knockdown and TIM-overexpressing A549 cells were constructed. Cancer behaviors in vitro were determined by colony formation, EdU proliferation, wound healing, transwell migration, flow cytometer, and CCK8 assays. Immunofluorescence, pathology examinations, and targeted metabolomics were also used in tumor cells and tissues. mCherry-GFP-LC3 plasmid was used to detect autophagic flux. RESULTS We found for the first time that compared to normal ad libitum feeding, 6-h TRF inhibited lung cancer progression and reprogrammed the rhythms of metabolites or genes involved in glycolysis and the circadian rhythm in tumors. After TRF intervention, only timeless (TIM) gene among five lung cancer-associated clock genes was found to consistently align rhythm of tumor cells to that of tumor tissues. Further, we demonstrated that the anti-tumor effect upon TRF was partially mediated by the rhythmic downregulation of the TIM and the subsequent activation of autophagy. Combining TRF with TIM inhibition further enhanced the anti-tumor effect, comparable to treatment efficacy of chemotherapy in xenograft model. CONCLUSIONS Six-hour TRF inhibits lung cancer progression and reshapes circadian metabolism, which is partially mediated by the rhythmic downregulation of the TIM and the subsequent upregulation of autophagy.
Collapse
Affiliation(s)
- Dan Shi
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P.R. China.
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, P.R. China.
- Research Center for Environment and Population Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China.
- Nutrition Innovation Platform-Sichuan and Chongqing, School of Public Health, Chongqing Medical University, Chongqing, China.
| | - Gaofeng Fang
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P.R. China
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Qianyao Chen
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P.R. China
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Jianling Li
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P.R. China
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Xiongzhong Ruan
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P.R. China.
| | - Xuemei Lian
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P.R. China.
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, P.R. China.
| |
Collapse
|
13
|
Wang K, Wang L, Chen X, Gu J, Cheng X. The role of N 6-methyladenosine RNA modification in platinum resistance. Epigenomics 2023; 15:1221-1232. [PMID: 38009226 DOI: 10.2217/epi-2023-0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023] Open
Abstract
N6-methyladenosine (m6A) RNA methylation, a dynamic regulator of transcript expression, plays a pivotal role in cancer by influencing diverse mRNA processes, including nuclear export, splicing, translation and decay. It intersects with cancer biology, impacting progression, treatment sensitivity and prognosis. Platinum-based compounds are essential in cancer treatment, while intrinsic or acquired resistance poses a formidable challenge, limiting therapeutic efficacy. Recent breakthroughs have established a direct association between m6A RNA methylation and platinum resistance in various cancer types. This review summarized related studies, aiming to provide profound insights into the interplay between m6A-associated regulation and platinum-resistance mechanisms in cancer. It explores therapeutic approaches, including personalized treatments based on m6A profiles, guiding future research to enhance clinical strategies for oncological prognostic outcomes.
Collapse
Affiliation(s)
- Kai Wang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Department of Obstetrics & Gynecology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Lingfang Wang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Xiaojing Chen
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Jiaxin Gu
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| |
Collapse
|
14
|
Khazaei Y, Basi A, Fernandez ML, Foudazi H, Bagherzadeh R, Shidfar F. The effects of synbiotics supplementation on reducing chemotherapy-induced side effects in women with breast cancer: a randomized placebo-controlled double-blind clinical trial. BMC Complement Med Ther 2023; 23:339. [PMID: 37752516 PMCID: PMC10521476 DOI: 10.1186/s12906-023-04165-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND The prevalence of breast cancer and its mortality rate are increasing rapidly among women worldwide. On other hand, the courses of chemotherapy as the main treatment for these patients are too much exhaustive and annoying. This study was designed to evaluate the use of synbiotics (probiotics + prebiotics) supplementation as a safe and inexpensive adjuvant treatment in reducing common chemotherapy side effects in women with breast cancer. METHODS The current study was conducted on 67 women with definitive diagnosis of breast cancer who were hospitalized to receive one-day chemotherapy sessions, and met the inclusion criteria. The patients were randomly allocated to the intervention or control group to receive synbiotics or placebo, respectively. They received oral consumption of synbiotics supplements twice a day for 8 weeks. The primary outcome was the changes in severity or experience of chemotherapy complication, analyzed by intention to treat (ITT). The instruments included 7 validated questionnaires which were used to assess chemotherapy complications in the initiation, 4 weeks and 8 weeks after intervention. Dietary intake was measured by 24-h dietary recall at the beginning, week 4 and week 8. Data were analyzed by SPSS software version 24. P-value < 0.05 was considered as statistically significant. RESULTS 67 breast cancer patients participated in the study. 8 weeks after intervention and adjusting the confounders, the severity of chemotherapy complications including unnormal defecation (P = 0.005) and fatigue (P < 0/001) decreased significantly in the synbiotics group compared to the placebo group. Furthermore, nausea/vomiting (P = 0.015), and anorexia (P < 0.001) were decreased at the end of the study compared to the first visit, but it was not statistically significant compared to the placebo group. CONCLUSIONS Synbiotics supplementation during chemotherapy can potentially reduce the severity of fatigue and abnormal defecation. It can help reduce anorexia and nausea/vomiting. TRIAL REGISTRATION This study was registered in the Iranian Registry of Clinical Trials (IRCT) (registered code: IRCT20091114002709N56) (date of registration: 5/5/2021). Direct link to the trial page: https://www.irct.ir/trial/54559 .
Collapse
Affiliation(s)
- Yasaman Khazaei
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Ali Basi
- Department of Hematology Oncology, Iran University of Medical Sciences, Tehran, Iran
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Hossein Foudazi
- Department of Radiation and Oncology, Shahid Fayaz-Bakhsh Hospital, Tehran, Iran
| | - Rafat Bagherzadeh
- English Department, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran.
| |
Collapse
|
15
|
Khazaei Y, Basi A, Fernandez ML, Foudazi H, Bagherzadeh R, Shidfar F. The effects of synbiotics supplementation on reducing chemotherapy-induced side effects in women with breast cancer: a randomized placebo-controlled double-blind clinical trial. BMC Complement Med Ther 2023; 23:339. [PMID: 37752516 DOI: 10.1186/s12906-023-04165-8.pmid:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/12/2023] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The prevalence of breast cancer and its mortality rate are increasing rapidly among women worldwide. On other hand, the courses of chemotherapy as the main treatment for these patients are too much exhaustive and annoying. This study was designed to evaluate the use of synbiotics (probiotics + prebiotics) supplementation as a safe and inexpensive adjuvant treatment in reducing common chemotherapy side effects in women with breast cancer. METHODS The current study was conducted on 67 women with definitive diagnosis of breast cancer who were hospitalized to receive one-day chemotherapy sessions, and met the inclusion criteria. The patients were randomly allocated to the intervention or control group to receive synbiotics or placebo, respectively. They received oral consumption of synbiotics supplements twice a day for 8 weeks. The primary outcome was the changes in severity or experience of chemotherapy complication, analyzed by intention to treat (ITT). The instruments included 7 validated questionnaires which were used to assess chemotherapy complications in the initiation, 4 weeks and 8 weeks after intervention. Dietary intake was measured by 24-h dietary recall at the beginning, week 4 and week 8. Data were analyzed by SPSS software version 24. P-value < 0.05 was considered as statistically significant. RESULTS 67 breast cancer patients participated in the study. 8 weeks after intervention and adjusting the confounders, the severity of chemotherapy complications including unnormal defecation (P = 0.005) and fatigue (P < 0/001) decreased significantly in the synbiotics group compared to the placebo group. Furthermore, nausea/vomiting (P = 0.015), and anorexia (P < 0.001) were decreased at the end of the study compared to the first visit, but it was not statistically significant compared to the placebo group. CONCLUSIONS Synbiotics supplementation during chemotherapy can potentially reduce the severity of fatigue and abnormal defecation. It can help reduce anorexia and nausea/vomiting. TRIAL REGISTRATION This study was registered in the Iranian Registry of Clinical Trials (IRCT) (registered code: IRCT20091114002709N56) (date of registration: 5/5/2021). Direct link to the trial page: https://www.irct.ir/trial/54559 .
Collapse
Affiliation(s)
- Yasaman Khazaei
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Ali Basi
- Department of Hematology Oncology, Iran University of Medical Sciences, Tehran, Iran
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Hossein Foudazi
- Department of Radiation and Oncology, Shahid Fayaz-Bakhsh Hospital, Tehran, Iran
| | - Rafat Bagherzadeh
- English Department, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran.
| |
Collapse
|
16
|
Shabkhizan R, Haiaty S, Moslehian MS, Bazmani A, Sadeghsoltani F, Saghaei Bagheri H, Rahbarghazi R, Sakhinia E. The Beneficial and Adverse Effects of Autophagic Response to Caloric Restriction and Fasting. Adv Nutr 2023; 14:1211-1225. [PMID: 37527766 PMCID: PMC10509423 DOI: 10.1016/j.advnut.2023.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/04/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023] Open
Abstract
Each cell is equipped with a conserved housekeeping mechanism, known as autophagy, to recycle exhausted materials and dispose of injured organelles via lysosomal degradation. Autophagy is an early-stage cellular response to stress stimuli in both physiological and pathological situations. It is thought that the promotion of autophagy flux prevents host cells from subsequent injuries by removing damaged organelles and misfolded proteins. As a correlate, the modulation of autophagy is suggested as a therapeutic approach in diverse pathological conditions. Accumulated evidence suggests that intermittent fasting or calorie restriction can lead to the induction of adaptive autophagy and increase longevity of eukaryotic cells. However, prolonged calorie restriction with excessive autophagy response is harmful and can stimulate a type II autophagic cell death. Despite the existence of a close relationship between calorie deprivation and autophagic response in different cell types, the precise molecular mechanisms associated with this phenomenon remain unclear. Here, we aimed to highlight the possible effects of prolonged and short-term calorie restriction on autophagic response and cell homeostasis.
Collapse
Affiliation(s)
- Roya Shabkhizan
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Sadat Moslehian
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Bazmani
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Student Committee Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ebrahim Sakhinia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Tang D, Tang Q, Huang W, Zhang Y, Tian Y, Fu X. Fasting: From Physiology to Pathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204487. [PMID: 36737846 PMCID: PMC10037992 DOI: 10.1002/advs.202204487] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/06/2023] [Indexed: 06/18/2023]
Abstract
Overnutrition is a risk factor for various human diseases, including neurodegenerative diseases, metabolic disorders, and cancers. Therefore, targeting overnutrition represents a simple but attractive strategy for the treatment of these increasing public health threats. Fasting as a dietary intervention for combating overnutrition has been extensively studied. Fasting has been practiced for millennia, but only recently have its roles in the molecular clock, gut microbiome, and tissue homeostasis and function emerged. Fasting can slow aging in most species and protect against various human diseases, including neurodegenerative diseases, metabolic disorders, and cancers. These centuried and unfading adventures and explorations suggest that fasting has the potential to delay aging and help prevent and treat diseases while minimizing side effects caused by chronic dietary interventions. In this review, recent animal and human studies concerning the role and underlying mechanism of fasting in physiology and pathology are summarized, the therapeutic potential of fasting is highlighted, and the combination of pharmacological intervention and fasting is discussed as a new treatment regimen for human diseases.
Collapse
Affiliation(s)
- Dongmei Tang
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuan610041China
| | - Qiuyan Tang
- Neurology Department of Integrated Traditional Chinese and Western Medicine, School of Clinical MedicineChengdu University of Traditional Chinese MedicineChengduSichuan610075China
| | - Wei Huang
- West China Centre of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineWest China‐Liverpool Biomedical Research CentreWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Yuwei Zhang
- Division of Endocrinology and MetabolismWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Yan Tian
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuan610041China
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuan610041China
| |
Collapse
|
18
|
Abstract
Experimental trials in organisms ranging from yeast to humans have shown that various forms of reducing food intake (caloric restriction) appear to increase both overall and healthy lifespan, delaying the onset of disease and slowing the progression of biomarkers of aging. The gut microbiota is considered one of the key environmental factors strongly contributing to the regulation of host health. Perturbations in the composition and activity of the gut microbiome are thought to be involved in the emergence of multiple diseases. Indeed, many studies investigating gut microbiota have been performed and have shown strong associations between specific microorganisms and metabolic diseases including overweight, obesity, and type 2 diabetes mellitus as well as specific gastrointestinal disorders, neurodegenerative diseases, and even cancer. Dietary interventions known to reduce inflammation and improve metabolic health are potentiated by prior fasting. Inversely, birth weight differential host oxidative phosphorylation response to fasting implies epigenetic control of some of its effector pathways. There is substantial evidence for the efficacy of fasting in improving insulin signaling and blood glucose control, and in reducing inflammation, conditions for which, additionally, the gut microbiota has been identified as a site of both risk and protective factors. Accordingly, human gut microbiota, both in symbiont and pathobiont roles, have been proposed to impact and mediate some health benefits of fasting and could potentially affect many of these diseases. While results from small-N studies diverge, fasting consistently enriches widely recognized anti-inflammatory gut commensals such as Faecalibacterium and other short-chain fatty acid producers, which likely mediates some of its health effects through immune system and barrier function impact.
Collapse
Affiliation(s)
- Sofia K Forslund
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Structural and Computational Biology Unit, EMBL, Heidelberg, Germany
| |
Collapse
|
19
|
Obesity is associated with early recurrence on breast cancer patients that achieved pathological complete response to neoadjuvant chemotherapy. Sci Rep 2022; 12:21145. [PMID: 36477462 PMCID: PMC9729290 DOI: 10.1038/s41598-022-25043-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Pathological complete response (pCR) after neoadjuvant chemotherapy (NCT) is associated with good long-term prognosis in breast cancer (BC) patients. However, some patients still recur and eventually die from this disease. For years, clinical stage at diagnosis has been consistently linked to recurrence and survival in the pCR setting. Herein, we aimed to identify other potential predictors of recurrence and survival in patients that achieved pCR. We performed a retrospective analysis of patients diagnosed between 2011 and 2020 in our center. We calculated overall survival (OS), invasive disease-free survival (IDFS), distant disease-free survival (DDFS), and BC-specific survival (BCSS). Among the 241 patients included into our study 36% were obese (Body Mass Index (BMI) > 29.9 kg/m2) and 47% were stage III. Multivariate analysis confirmed that obesity was a significant risk factor associated with early recurrence and poorer survival in these patients. In summary, obesity and clinical stage predict early recurrence and poorer survival in patients that achieved pCR after NCT. Pending further investigation and based on our findings we speculate that weight management could be beneficial for this subset of patients. To our knowledge, this is the first Latin American report linking obesity and recurrence within this setting.
Collapse
|
20
|
Liu L, Chen W, Wu D, Hu F. Metabolic Efficacy of Time-Restricted Eating in Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Clin Endocrinol Metab 2022; 107:3428-3441. [PMID: 36190980 DOI: 10.1210/clinem/dgac570] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Time-restricted eating (TRE), which restricts food intake to a limited duration of the day, is a key regimen of intermittent fasting. OBJECTIVE The aim of our study was to provide an up-to-date meta-analysis and systematic review to evaluate the efficacy of TRE on weight loss and other metabolic-related parameters in adults. METHODS We searched PubMed, EMBASE, and the Cochrane Library for relevant studies published before February 26, 2022. Study duration of TRE was at least 4 weeks. Body weight and other metabolic-related continuous parameters were described as weighted mean difference (WMD) with 95% CI. RESULTS Seventeen randomized controlled trials involving 899 participants were analyzed. The pooled meta-analysis has shown that TRE contributed to a significant decrease in body weight with a WMD of -1.60 kg (95% CI -2.27 to -0.93) and fat mass with WMD -1.48 kg (95% CI -1.59 to -1.38). Subgroup analysis showed that TRE could reduce body weight and fat mass especially in overweight participants with WMD -1.43 kg (95% CI -2.05 to -0.81) and -1.56 kg (95% CI -1.67 to -1.44), respectively. TRE also showed beneficial effects on the lipid spectrum in overweight participants, including decreased levels of triglyceride (WMD -12.71 mg/dL, 95% CI -24.9 to -0.52), total cholesterol (WMD -6.45 mg/dL, 95% CI -7.40 to -5.49), and low-density lipoprotein cholesterol (WMD -7.0 mg/dL, 95% CI -9.74 to -4.25). However, compared with control, TRE had no significant effects on waist circumference, body mass index, glycosylated hemoglobin, or blood pressure. CONCLUSION This updated meta-analysis found that TRE may be an effective approach to improve the metabolic state of nonobese subjects, especially in overweight participants.
Collapse
Affiliation(s)
- Lili Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Wei Chen
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Dan Wu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Fang Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| |
Collapse
|
21
|
Minciuna I, van Kleef LA, Stefanescu H, Procopet B. Is Fasting Good When One Is at Risk of Liver Cancer? Cancers (Basel) 2022; 14:5084. [PMID: 36291868 PMCID: PMC9600146 DOI: 10.3390/cancers14205084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC), one of the leading causes of cancer-related deaths worldwide, is a multistep process that usually develops in the background of cirrhosis, but also in a non-cirrhotic state in patients with non-alcoholic fatty liver disease (NAFLD) or viral hepatis. Emerging evidence suggests that intermittent fasting can reduce the risk of cancer development and could improve response and tolerance to treatment through the metabolic and hormonal adaptations induced by the low energy availability that finally impairs cancer cells' adaptability, survival and growth. The current review will outline the beneficial effects of fasting in NAFLD/NASH patients and the possible mechanisms that can prevent HCC development, including circadian clock re-synchronization, with a special focus on the possibility of applying this dietary intervention to cirrhotic patients.
Collapse
Affiliation(s)
- Iulia Minciuna
- Regional Institute of Gastroenterology and Hepatology Octavian Fodor, 400394 Cluj-Napoca, Romania
- 3rd Medical Department, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania
| | - Laurens A. van Kleef
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Horia Stefanescu
- Regional Institute of Gastroenterology and Hepatology Octavian Fodor, 400394 Cluj-Napoca, Romania
| | - Bogdan Procopet
- Regional Institute of Gastroenterology and Hepatology Octavian Fodor, 400394 Cluj-Napoca, Romania
- 3rd Medical Department, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania
| |
Collapse
|
22
|
Bossi P, De Luca R, Ciani O, D’Angelo E, Caccialanza R. Malnutrition management in oncology: An expert view on controversial issues and future perspectives. Front Oncol 2022; 12:910770. [PMID: 36276153 PMCID: PMC9579941 DOI: 10.3389/fonc.2022.910770] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/13/2022] [Indexed: 12/05/2022] Open
Abstract
Cancer and anticancer treatments can lead to several negative side effects, including malnutrition. Despite the recognized need for adequate nutritional support in cancer patients, in daily clinical practice, nutrition is still not considered one of the first aspects to be considered. Malnutrition negatively affects the clinical outcomes, treatment response, and overall survival of cancer patients. In this study, three of the most controversial issues related to malnutrition, which emerged during an Italian Consensus Conference, were addressed specifically for patients with head and neck as well as gastrointestinal cancer. The timing of nutritional evaluation and intervention, extension of the Enhanced Recovery after Surgery (ERAS®) protocols, and cost-effectiveness of nutritional interventions have been considered. This study aimed to illustrate the state-of-the art of each issue and hypothesize future perspectives and actions to be taken, trying to suggest a new nutritional management model for cancer patients in Italy that overcomes the critical issues encountered. Of note, the timely diagnosis of nutritional issue appears to be essential to ensure the correct management of malnourished cancer patients as well as those who are at high risk of malnutrition. Standardized protocols, screening tests, and the inclusion of nutritional parameters in patient medical records would help to achieve good clinical outcomes. Finally, appropriate nutritional support is also associated with cost savings, and it seems necessary to promote its clinical and economic value to obtain improvements in both outcomes and management costs.
Collapse
Affiliation(s)
- Paolo Bossi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Università degli Studi di Brescia, Brescia, Italy
| | - Raffaele De Luca
- Department of Surgical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II”, Bari, Italy
- *Correspondence: Raffaele De Luca,
| | - Oriana Ciani
- Centre for Research on Health and Social Care Management (CERGAS), Scuola di Direzione Aziendale (SDA) Bocconi School of Management, Bocconi University, Milan, Italy
| | - Elisa D’Angelo
- Radiotherapy Unit, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
23
|
Tiwari S, Sapkota N, Han Z. Effect of fasting on cancer: A narrative review of scientific evidence. Cancer Sci 2022; 113:3291-3302. [PMID: 35848874 PMCID: PMC9530862 DOI: 10.1111/cas.15492] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022] Open
Abstract
Emerging evidence suggests that fasting could play a key role in cancer treatment by fostering conditions that limit cancer cells' adaptability, survival, and growth. Fasting could increase the effectiveness of cancer treatments and limit adverse events. Yet, we lack an integrated mechanistic model for how these two complicated systems interact, limiting our ability to understand, prevent, and treat cancer using fasting. Here, we review recent findings at the interface of oncology and fasting metabolism, with an emphasis on human clinical studies of intermittent fasting. We recommend combining prolonged periodic fasting with a standard conventional therapeutic approach to promote cancer-free survival, treatment efficacy and reduce side effects in cancer patients.
Collapse
Affiliation(s)
- Sagun Tiwari
- Department of Neurology and RehabilitationSeventh People's Hospital of Shanghai University of TCMShanghaiChina
- Shanghai University of TCMShanghaiChina
- Life Care HospitalBagmatiNepal
| | - Namrata Sapkota
- University of Chinese Academy of SciencesBeijingChina
- Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
- Net Fresh HospitalBagmatiNepal
| | - Zhenxiang Han
- Department of Neurology and RehabilitationSeventh People's Hospital of Shanghai University of TCMShanghaiChina
| |
Collapse
|
24
|
Pereira IC, Mascarenhas IF, Capetini VC, Ferreira PMP, Rogero MM, Torres-Leal FL. Cellular reprogramming, chemoresistance, and dietary interventions in breast cancer. Crit Rev Oncol Hematol 2022; 179:103796. [PMID: 36049616 DOI: 10.1016/j.critrevonc.2022.103796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/16/2022] [Accepted: 08/21/2022] [Indexed: 10/31/2022] Open
Abstract
Breast cancer (BC) diagnosis has been associated with significant risk factors, including family history, late menopause, obesity, poor eating habits, and alcoholism. Despite the advances in the last decades regarding cancer treatment, some obstacles still hinder the effectiveness of therapy. For example, chemotherapy resistance is common in locally advanced or metastatic cancer, reducing treatment options and contributing to mortality. In this review, we provide an overview of BC metabolic changes, including the impact of restrictive diets associated with chemoresistance, the therapeutic potential of the diet on tumor progression, pathways related to metabolic health in oncology, and perspectives on the future in the area of oncological nutrition.
Collapse
Affiliation(s)
- Irislene Costa Pereira
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil; Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Isabele Frazão Mascarenhas
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | - Paulo Michel Pinheiro Ferreira
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, Sao Paulo, Brazil
| | - Francisco Leonardo Torres-Leal
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil; Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil.
| |
Collapse
|
25
|
Isaac-Lam MF, DeMichael KM. Calorie restriction and breast cancer treatment: a mini-review. J Mol Med (Berl) 2022; 100:1095-1109. [PMID: 35760911 DOI: 10.1007/s00109-022-02226-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 06/02/2022] [Accepted: 06/10/2022] [Indexed: 12/11/2022]
Abstract
Calorie restriction (CR), referred to as a reduction in dietary calorie intake without malnutrition, has been demonstrated to be a safe way to extend longevity of yeast, worms, and laboratory animals, and to decrease the risk factors in age-related diseases including cancer in humans. Pre-clinical studies in animal models demonstrated that CR may enhance the efficacy of chemotherapy, radiation therapy, and immunotherapy during breast cancer treatment. Reduced calorie intake ameliorates risk factors and delays the onset of cancer by altering metabolism and fostering health-enhancing characteristics including increased autophagy and insulin sensitivity, and decreased blood glucose levels, inflammation, angiogenesis, and growth factor signaling. CR is not a common protocol implemented by medical practitioners to the general public due to the lack of substantial clinical studies. Future research and clinical trials are urgently needed to understand fully the biochemical basis of CR or CR mimetics to support its benefits. Here, we present a mini-review of research studies integrating CR as an adjuvant to chemotherapy, radiation therapy, or immunotherapy during breast cancer treatment.
Collapse
Affiliation(s)
- Meden F Isaac-Lam
- Department of Chemistry and Physics, Purdue University Northwest, Westville, IN, 46391, USA.
| | - Kelly M DeMichael
- Department of Chemistry and Physics, Purdue University Northwest, Westville, IN, 46391, USA
| |
Collapse
|
26
|
Bradey AL, Fitter S, Duggan J, Wilczek V, Williams CMD, Cheney EA, Noll JE, Tangseefa P, Panagopoulos V, Zannettino ACW. Calorie restriction has no effect on bone marrow tumour burden in a Vk*MYC transplant model of multiple myeloma. Sci Rep 2022; 12:13128. [PMID: 35908046 PMCID: PMC9338941 DOI: 10.1038/s41598-022-17403-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022] Open
Abstract
Multiple myeloma (MM) is an incurable haematological malignancy, caused by the uncontrolled proliferation of plasma cells within the bone marrow (BM). Obesity is a known risk factor for MM, however, few studies have investigated the potential of dietary intervention to prevent MM progression. Calorie restriction (CR) is associated with many health benefits including reduced cancer incidence and progression. To investigate if CR could reduce MM progression, dietary regimes [30% CR, normal chow diet (NCD), or high fat diet (HFD)] were initiated in C57BL/6J mice. Diet-induced changes were assessed, followed by inoculation of mice with Vk*MYC MM cells (Vk14451-GFP) at 16 weeks of age. Tumour progression was monitored by serum paraprotein, and at endpoint, BM and splenic tumour burden was analysed by flow cytometry. 30% CR promoted weight loss, improved glucose tolerance, increased BM adiposity and elevated serum adiponectin compared to NCD-fed mice. Despite these metabolic changes, CR had no significant effect on serum paraprotein levels. Furthermore, endpoint analysis found that dietary changes were insufficient to affect BM tumour burden, however, HFD resulted in an average two-fold increase in splenic tumour burden. Overall, these findings suggest diet-induced BM changes may not be key drivers of MM progression in the Vk14451-GFP transplant model of myeloma.
Collapse
Affiliation(s)
- Alanah L Bradey
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stephen Fitter
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Jvaughn Duggan
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Vicki Wilczek
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Connor M D Williams
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Emma Aj Cheney
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Jacqueline E Noll
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Pawanrat Tangseefa
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Vasilios Panagopoulos
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia. .,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia.
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia.,Department of Haematology, Royal Adelaide Hospital, Adelaide, Australia.,Central Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
27
|
Jawarneh S, Talib WH. Combination of Ashwagandha Water Extract and Intermittent Fasting as a Therapy to Overcome Cisplatin Resistance in Breast Cancer: An in vitro and in vivo Study. Front Nutr 2022; 9:863619. [PMID: 35859750 PMCID: PMC9290527 DOI: 10.3389/fnut.2022.863619] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is considered a universal public health dilemma in women. Due to the high toxicity and low selectivity of conventional anticancer therapies, there is a growing trend of using plant-derived natural products in cancer prevention and therapy. Ashwagandha (Withania somnifera, WS) has been used in the Mediterranean region and Ayurvedic medicine for millennia as a functional food and a medicinal plant with anticancer activity. Besides, intermittent fasting (IF) has been engaged recently in cancer treatment. Hence, the combination of WS and IF provides possible solutions to treat cancer and reduce chemoresistance when combined with chemotherapy. In this study, WS root (WSR), IF, and cisplatin were tested on cisplatin-sensitive (EMT6/P) and cisplatin-resistant (EMT6/CPR) mouse mammary cell lines. The phytochemical content of the WSR extract was analyzed using liquid chromatography–mass spectrometry (LC-MS) analysis. Antiproliferative and apoptotic effects were assessed for WSR extract, cisplatin, and their combination in vitro using [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] (MTT) and caspase-3 assays. An in vivo study was used to assess the effect of WSR extract, IF, cisplatin, and their combinations in mice inculcated with EMT6/P and EMT6/CPR cells. The safety profile was also investigated using liver enzymes and creatinine assays. In vitro, WSR extract and cisplatin had a synergistic effect in both cell lines. The same combination induced an apoptotic effect higher than the single treatment in both cell lines. In vivo, several combinations of WSR extract, IF, or cisplatin caused significant tumor size reduction and improved the cure rate in mice implanted with EMT6/P and EMT6/CPR cell lines. IF-treated groups showed a significant reduction in serum glucose and an elevation in β-hydroxybutyrate (BHB) levels. In the safety profile, WSR extract, IF, and their combinations were safe. Overall, the combination of WSR extract and IF provides a promising solution for breast cancer treatment besides cisplatin by reducing the proliferation of cancer cells through induction of apoptosis. Moreover, they minimize cisplatin toxicity to the liver and kidney.
Collapse
|
28
|
Ndembe G, Intini I, Perin E, Marabese M, Caiola E, Mendogni P, Rosso L, Broggini M, Colombo M. LKB1: Can We Target an Hidden Target? Focus on NSCLC. Front Oncol 2022; 12:889826. [PMID: 35646638 PMCID: PMC9131655 DOI: 10.3389/fonc.2022.889826] [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: 03/04/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
LKB1 (liver kinase B1) is a master regulator of several processes such as metabolism, proliferation, cell polarity and immunity. About one third of non-small cell lung cancers (NSCLCs) present LKB1 alterations, which almost invariably lead to protein loss, resulting in the absence of a potential druggable target. In addition, LKB1-null tumors are very aggressive and resistant to chemotherapy, targeted therapies and immune checkpoint inhibitors (ICIs). In this review, we report and comment strategies that exploit peculiar co-vulnerabilities to effectively treat this subgroup of NSCLCs. LKB1 loss leads to an enhanced metabolic avidity, and treatments inducing metabolic stress were successful in inhibiting tumor growth in several preclinical models. Biguanides, by compromising mitochondria and reducing systemic glucose availability, and the glutaminase inhibitor telaglenastat (CB-839), inhibiting glutamate production and reducing carbon intermediates essential for TCA cycle progression, have provided the most interesting results and entered different clinical trials enrolling also LKB1-null NSCLC patients. Nutrient deprivation has been investigated as an alternative therapeutic intervention, giving rise to interesting results exploitable to design specific dietetic regimens able to counteract cancer progression. Other strategies aimed at targeting LKB1-null NSCLCs exploit its pivotal role in modulating cell proliferation and cell invasion. Several inhibitors of LKB1 downstream proteins, such as mTOR, MEK, ERK and SRK/FAK, resulted specifically active on LKB1-mutated preclinical models and, being molecules already in clinical experimentation, could be soon proposed as a specific therapy for these patients. In particular, the rational use in combination of these inhibitors represents a very promising strategy to prevent the activation of collateral pathways and possibly avoid the potential emergence of resistance to these drugs. LKB1-null phenotype has been correlated to ICIs resistance but several studies have already proposed the mechanisms involved and potential interventions. Interestingly, emerging data highlighted that LKB1 alterations represent positive determinants to the new KRAS specific inhibitors response in KRAS co-mutated NSCLCs. In conclusion, the absence of the target did not block the development of treatments able to hit LKB1-mutated NSCLCs acting on several fronts. This will give patients a concrete chance to finally benefit from an effective therapy.
Collapse
Affiliation(s)
- Gloriana Ndembe
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilenia Intini
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisa Perin
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisa Caiola
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Paolo Mendogni
- Thoracic Surgery and Lung Transplantation Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplantation Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marika Colombo
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
29
|
Short-Term Fasting Synergizes with Solid Cancer Therapy by Boosting Antitumor Immunity. Cancers (Basel) 2022; 14:cancers14061390. [PMID: 35326541 PMCID: PMC8946179 DOI: 10.3390/cancers14061390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Stimulating our body’s own immune response to fight cancer is important for the success of cancer treatment in general. To further improve current cancer therapy, preclinical research shows that short-term fasting diets enhance cancer therapy efficacy, such as chemotherapy. Short-term fasting diets are low-caloric and low in protein for 3–5 days; they are usually done every couple of weeks. This review summarizes preclinical and clinical evidence of fasting diets synergizing with cancer therapy by boosting antitumor immunity. Abstract Short-term fasting (STF), using a low caloric, low protein fasting mimicking diet (FMD), appears to be a promising strategy to enhance chemotherapy-based cancer efficacy, while potentially alleviating toxicity. Preclinical results suggest that enhanced tumor immunity and decreased growth signaling, via lowering of circulating insulin and insulin growth factor 1 (IGF-1) levels form the potential underlying mechanisms. STF may boost anti-tumor responses by promoting tumor immunogenicity and decreasing local immunosuppression. These findings warrant further studies focused on the combination of STF, not only with chemotherapy, but also with immunotherapy to evaluate the full range of benefits of STF in cancer treatment. Here, we delineate the underlying anticancer mechanisms of fasting. We summarize preclinical evidence of STF boosting antitumor immunity and alleviating immunosuppression, as well as the clinical findings reporting the immunomodulatory effects of STF during various cancer treatments, including immunotherapy.
Collapse
|
30
|
Zhou L, Zhang Z, Nice E, Huang C, Zhang W, Tang Y. Circadian rhythms and cancers: the intrinsic links and therapeutic potentials. J Hematol Oncol 2022; 15:21. [PMID: 35246220 PMCID: PMC8896306 DOI: 10.1186/s13045-022-01238-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The circadian rhythm is an evolutionarily conserved time-keeping system that comprises a wide variety of processes including sleep-wake cycles, eating-fasting cycles, and activity-rest cycles, coordinating the behavior and physiology of all organs for whole-body homeostasis. Acute disruption of circadian rhythm may lead to transient discomfort, whereas long-term irregular circadian rhythm will result in the dysfunction of the organism, therefore increasing the risks of numerous diseases especially cancers. Indeed, both epidemiological and experimental evidence has demonstrated the intrinsic link between dysregulated circadian rhythm and cancer. Accordingly, a rapidly increasing understanding of the molecular mechanisms of circadian rhythms is opening new options for cancer therapy, possibly by modulating the circadian clock. In this review, we first describe the general regulators of circadian rhythms and their functions on cancer. In addition, we provide insights into the mechanisms underlying how several types of disruption of the circadian rhythm (including sleep-wake, eating-fasting, and activity-rest) can drive cancer progression, which may expand our understanding of cancer development from the clock perspective. Moreover, we also summarize the potential applications of modulating circadian rhythms for cancer treatment, which may provide an optional therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Edouard Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Acupuncture and Chronobiology Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
31
|
Bariwal J, Ma H, Altenberg GA, Liang H. Nanodiscs: a versatile nanocarrier platform for cancer diagnosis and treatment. Chem Soc Rev 2022; 51:1702-1728. [PMID: 35156110 DOI: 10.1039/d1cs01074c] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer therapy is a significant challenge due to insufficient drug delivery to the cancer cells and non-selective killing of healthy cells by most chemotherapy agents. Nano-formulations have shown great promise for targeted drug delivery with improved efficiency. The shape and size of nanocarriers significantly affect their transport inside the body and internalization into the cancer cells. Non-spherical nanoparticles have shown prolonged blood circulation half-lives and higher cellular internalization frequency than spherical ones. Nanodiscs are desirable nano-formulations that demonstrate enhanced anisotropic character and versatile functionalization potential. Here, we review the recent development of theranostic nanodiscs for cancer mitigation ranging from traditional lipid nanodiscs encased by membrane scaffold proteins to newer nanodiscs where either the membrane scaffold proteins or the lipid bilayers themselves are replaced with their synthetic analogues. We first discuss early cancer detection enabled by nanodiscs. We then explain different strategies that have been explored to carry a wide range of payloads for chemotherapy, cancer gene therapy, and cancer vaccines. Finally, we discuss recent progress on organic-inorganic hybrid nanodiscs and polymer nanodiscs that have the potential to overcome the inherent instability problem of lipid nanodiscs.
Collapse
Affiliation(s)
- Jitender Bariwal
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Hairong Ma
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Hongjun Liang
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
32
|
Vidoni C, Ferraresi A, Esposito A, Maheshwari C, Dhanasekaran DN, Mollace V, Isidoro C. Calorie Restriction for Cancer Prevention and Therapy: Mechanisms, Expectations, and Efficacy. J Cancer Prev 2021; 26:224-236. [PMID: 35047448 PMCID: PMC8749320 DOI: 10.15430/jcp.2021.26.4.224] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/18/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer is one of the most frequently diagnosed diseases, and despite the continuous efforts in searching for new and more effective treatments, its morbidity and mortality remain a significant health problem worldwide. Calorie restriction, a dietary manipulation that consists in a reduction of the calorie intake, is gaining attention as a potential adjuvant intervention for preventing and/or fighting cancer. Several forms of energy reduction intake, which includes caloric restriction tout-court, dietary restrictions, and intermittent fasting, are being explored for their ability to prevent or slow down cancer progression. Additionally, another anti-cancer approach being under investigation relies on the use of nutraceuticals known as “Caloric Restriction Mimetics” that can provide caloric restriction-mediated benefits without subjecting the patients to a strict diet. Preclinical in vitro and in vivo studies consistently show that diet modifiers reducing the calorie have impact on tumor microenvironment and cancer metabolism, resulting in reduced growth and progression of cancer. Preliminary clinical studies show that patients subjected to a reduced nutrient/energy intake experience improved outcomes from chemo- and radiotherapy while better tolerating the side effects. Here, we review the state of the art on the therapeutic potential of calorie restriction and of caloric restriction mimetics in preventing or retarding tumor development by modulating a subset of cellular processes. The most recent clinical progresses with caloric restriction mimetics in the clinical practice are also discussed.
Collapse
Affiliation(s)
- Chiara Vidoni
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Andrea Esposito
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Chinmay Maheshwari
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Vincenzo Mollace
- Department of Health Sciences, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| |
Collapse
|
33
|
Anderson PM, Thomas SM, Sartoski S, Scott JG, Sobilo K, Bewley S, Salvador LK, Salazar-Abshire M. Strategies to Mitigate Chemotherapy and Radiation Toxicities That Affect Eating. Nutrients 2021; 13:nu13124397. [PMID: 34959948 PMCID: PMC8706251 DOI: 10.3390/nu13124397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Cancer and its therapy is commonly associated with a variety of side effects that impact eating behaviors that reduce nutritional intake. This review will outline potential causes of chemotherapy and radiation damage as well as approaches for the amelioration of the side effects of cancer during therapy. Methods: Information for clinicians, patients, and their caregivers about toxicity mitigation including nausea reduction, damage to epithelial structures such as skin and mucosa, organ toxicity, and education is reviewed. Results: How to anticipate, reduce, and prevent some toxicities encountered during chemotherapy and radiation is detailed with the goal to improve eating behaviors. Strategies for health care professionals, caregivers, and patients to consider include (a) the reduction in nausea and vomiting, (b) decreasing damage to the mucosa, (c) avoiding a catabolic state and muscle wasting (sarcopenia), and (d) developing therapeutic alliances with patients, caregivers, and oncologists. Conclusions: Although the reduction of side effects involves anticipatory guidance and proactive team effort (e.g., forward observation, electronic interactions, patient reported outcomes), toxicity reduction can be satisfying for not only the patient, but everyone involved in cancer care.
Collapse
Affiliation(s)
- Peter M. Anderson
- Pediatric Hematology/Oncology and Bone Marrow Transplant, Cleveland Clinic Children’s, Cleveland, OH 44195, USA; (S.M.T.); (S.S.); (K.S.); (S.B.)
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Correspondence: or ; Tel.: +216-445-7140 or +216-308-2706
| | - Stefanie M. Thomas
- Pediatric Hematology/Oncology and Bone Marrow Transplant, Cleveland Clinic Children’s, Cleveland, OH 44195, USA; (S.M.T.); (S.S.); (K.S.); (S.B.)
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Shauna Sartoski
- Pediatric Hematology/Oncology and Bone Marrow Transplant, Cleveland Clinic Children’s, Cleveland, OH 44195, USA; (S.M.T.); (S.S.); (K.S.); (S.B.)
- Department of Nursing, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jacob G. Scott
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Department of Radiation Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kaitlin Sobilo
- Pediatric Hematology/Oncology and Bone Marrow Transplant, Cleveland Clinic Children’s, Cleveland, OH 44195, USA; (S.M.T.); (S.S.); (K.S.); (S.B.)
- Department of Nursing, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Sara Bewley
- Pediatric Hematology/Oncology and Bone Marrow Transplant, Cleveland Clinic Children’s, Cleveland, OH 44195, USA; (S.M.T.); (S.S.); (K.S.); (S.B.)
- Peds Nutritional Services, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Laura K. Salvador
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX 77030, USA; (L.K.S.); (M.S.-A.)
| | - Maritza Salazar-Abshire
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX 77030, USA; (L.K.S.); (M.S.-A.)
- Department of Nursing Education, MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
34
|
Selvan SR, Brichetti JA, Thurber DB, Botting GM, Bertenshaw GP. Functional Profiling of Head and Neck/Esophageal Squamous Cell Carcinoma to Predict Cetuximab Response. Cancer Biother Radiopharm 2021. [PMID: 34846938 DOI: 10.1089/cbr.2021.0283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background: Cetuximab, an epidermal growth factor receptor (EGFR)-targeting antibody, remains the only Food and Drug Administration-approved targeted therapy for squamous cell carcinoma (SCC) of head and neck/esophagus. However, in clinical trials, cetuximab only benefited a subset of patients and frequently caused toxicity. Predicting which patients respond to cetuximab remains unsolved. The authors sought to identify predictive biomarkers in EGFR signaling and autophagy pathways, which may be impacted by cetuximab under certain treatment conditions. Methods: In vitro responses of SCC cell lines to cetuximab under various nutrient conditions were assessed by WST-8 growth assay. Functional profiles of several EGFR signaling biomarkers were investigated by Luminex-based assays and corroborated with immunoblots. Autophagy markers were analyzed with immunoblots. Results: In vitro growth response assays identified cetuximab responder and nonresponder cell lines. Optimal growth conditions and growth factors enhanced responses, and even reversed nonresponsiveness in some cell lines. Strong correlation was found between response in growth assays (reference assay) and dynamic changes in p-Erk1/2 and LC3-II (index assays). Conclusions: This study indicates that nutrient modification may enhance cetuximab response in SCC patients. Biomarker results strengthen the hypothesis that dynamic biomarkers can be used to predict patient response to cetuximab. Future studies are warranted to test in more complex samples including patient-derived tumor tissues.
Collapse
|
35
|
Aprile G, Basile D, Giaretta R, Schiavo G, La Verde N, Corradi E, Monge T, Agustoni F, Stragliotto S. The Clinical Value of Nutritional Care before and during Active Cancer Treatment. Nutrients 2021; 13:nu13041196. [PMID: 33916385 PMCID: PMC8065908 DOI: 10.3390/nu13041196] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
Malnutrition and muscle wasting are frequently reported in cancer patients, either linked to the tumor itself or caused by oncologic therapies. Understanding the value of nutritional care during cancer treatment remains crucial. In fact, cancer-associated sarcopenia plays a key role in determining higher rates of morbidity, mortality, treatment-induced toxicities, prolonged hospitalizations and reduced adherence to anticancer treatment, worsening quality of life and survival. Planning baseline screening to intercept nutritional troubles earlier, organizing timely reassessments, and providing adequate counselling and dietary support, healthcare professional may positively interfere with this process and improve patients' overall outcomes during the whole disease course. Several screening tools have been proposed for this purpose. Nutritional Risk Screening (NRS), Mini Nutritional Assessment (MNA), Patient Generated Subjective Global Assessment (PG-SGA) are the most common studied. Interestingly, second-level tools including skeletal muscle index (SMI) and bioelectric impedance analysis (BIA) provide a more precise assessment of body composition, even if they are more complex. However, nutritional assessment is not currently used in clinical practice and procedures must be standardized in order to improve the efficacy of standard chemotherapy, targeted agents or even checkpoint inhibitors that is potentially linked with the patients' nutritional status. In the present review, we will discuss about malnutrition and the importance of an early nutritional assessment during chemotherapy and treatment with novel checkpoint inhibitors, in order to prevent treatment-induced toxicities and to improve survival outcomes.
Collapse
Affiliation(s)
- Giuseppe Aprile
- Department of Oncology, AULSS8 Berica, 36100 Vicenza, Italy; (D.B.); (R.G.)
- Correspondence: ; Tel.: +39-0444753906
| | - Debora Basile
- Department of Oncology, AULSS8 Berica, 36100 Vicenza, Italy; (D.B.); (R.G.)
| | - Renato Giaretta
- Department of Oncology, AULSS8 Berica, 36100 Vicenza, Italy; (D.B.); (R.G.)
| | - Gessica Schiavo
- Clinical Nutritional Unit, AULSS8 Berica, 36100 Vicenza, Italy;
| | - Nicla La Verde
- Department of Oncology, PO Sacco, ASST Fatebenefratelli Sacco, 20131 Milano, Italy;
| | - Ettore Corradi
- Clinical Nutritional Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy;
| | - Taira Monge
- Clinical Nutrition, S. Giovanni Battista Hospital, 10126 Torino, Italy;
| | - Francesco Agustoni
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Silvia Stragliotto
- Department of Oncology, Istituto Oncologico Veneto—IRCCS, 31033 Padova, Italy;
| |
Collapse
|