1
|
Wan S, Zhou X, Xie F, Zhou F, Zhang L. Ketogenic diet and cancer: multidimensional exploration and research. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1010-1024. [PMID: 39821829 DOI: 10.1007/s11427-023-2637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/29/2024] [Indexed: 01/19/2025]
Abstract
The ketogenic diet (KD) has attracted attention in recent years for its potential anticancer effects. KD is a dietary structure of high fat, moderate protein, and extremely low carbohydrate content. Originally introduced as a treatment for epilepsy, KD has been widely applied in weight loss programs and the management of metabolic diseases. Previous studies have shown that KD can potentially inhibit the growth and spread of cancer by limiting energy supply to tumor cells, thereby inhibiting tumor angiogenesis, reducing oxidative stress in normal cells, and affecting cancer cell signaling and other processes. Moreover, KD has been shown to influence T-cell-mediated immune responses and inflammation by modulating the gut microbiota, enhance the efficacy of standard cancer treatments, and mitigate the complications of chemotherapy. However, controversies and uncertainties remain regarding the specific mechanisms and clinical effects of KD as an adjunctive therapy for cancer. Therefore, this review summarizes the existing research and explores the intricate relationships between KD and cancer treatment.
Collapse
Affiliation(s)
- Shiyun Wan
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, China.
| | - Long Zhang
- Life Sciences Institute and State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- Cancer Center Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Zuo Q, Yoo JY, Nelson ER, Sikora MJ, Riggins RB, Madak-Erdogan Z. Co-targeting of metabolism using dietary and pharmacologic approaches reduces breast cancer metastatic burden. NPJ Breast Cancer 2025; 11:3. [PMID: 39809806 PMCID: PMC11733225 DOI: 10.1038/s41523-024-00715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Patients with metastatic breast cancer face reduced quality of life and increased mortality rates, necessitating more effective anti-cancer strategies. Building on previous research that identified metastatic-niche-specific metabolic vulnerabilities, we investigated how a ketogenic diet enhances estrogen receptor (ER)-positive liver metastatic breast cancer's response to Fulvestrant (Fulv) treatment. Using in vitro cell lines and in vivo xenograft metastasis mouse models, we examined the molecular mechanisms of combining ER targeting with a ketogenic diet. We found that Fulv treatment downregulates the ketogenesis pathway enzyme OXCT1, leading to β-hydroxybutyrate accumulation and decreased tumor cell viability. We also explored interactions between glucose, palmitic acid, and β-hydroxybutyric acid. These findings establish the molecular basis and clinical potential of a ketogenic diet to enhance Fulv efficacy in patients with ER+ liver metastatic breast cancer, potentially improving survival outcomes and quality of life in this population.
Collapse
Affiliation(s)
- Qianying Zuo
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jin Young Yoo
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Matthew J Sikora
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca B Riggins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
3
|
Duraj T, Kalamian M, Zuccoli G, Maroon JC, D'Agostino DP, Scheck AC, Poff A, Winter SF, Hu J, Klement RJ, Hickson A, Lee DC, Cooper I, Kofler B, Schwartz KA, Phillips MCL, Champ CE, Zupec-Kania B, Tan-Shalaby J, Serfaty FM, Omene E, Arismendi-Morillo G, Kiebish M, Cheng R, El-Sakka AM, Pflueger A, Mathews EH, Worden D, Shi H, Cincione RI, Spinosa JP, Slocum AK, Iyikesici MS, Yanagisawa A, Pilkington GJ, Chaffee A, Abdel-Hadi W, Elsamman AK, Klein P, Hagihara K, Clemens Z, Yu GW, Evangeliou AE, Nathan JK, Smith K, Fortin D, Dietrich J, Mukherjee P, Seyfried TN. Clinical research framework proposal for ketogenic metabolic therapy in glioblastoma. BMC Med 2024; 22:578. [PMID: 39639257 PMCID: PMC11622503 DOI: 10.1186/s12916-024-03775-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, with a universally lethal prognosis despite maximal standard therapies. Here, we present a consensus treatment protocol based on the metabolic requirements of GBM cells for the two major fermentable fuels: glucose and glutamine. Glucose is a source of carbon and ATP synthesis for tumor growth through glycolysis, while glutamine provides nitrogen, carbon, and ATP synthesis through glutaminolysis. As no tumor can grow without anabolic substrates or energy, the simultaneous targeting of glycolysis and glutaminolysis is expected to reduce the proliferation of most if not all GBM cells. Ketogenic metabolic therapy (KMT) leverages diet-drug combinations that inhibit glycolysis, glutaminolysis, and growth signaling while shifting energy metabolism to therapeutic ketosis. The glucose-ketone index (GKI) is a standardized biomarker for assessing biological compliance, ideally via real-time monitoring. KMT aims to increase substrate competition and normalize the tumor microenvironment through GKI-adjusted ketogenic diets, calorie restriction, and fasting, while also targeting glycolytic and glutaminolytic flux using specific metabolic inhibitors. Non-fermentable fuels, such as ketone bodies, fatty acids, or lactate, are comparatively less efficient in supporting the long-term bioenergetic and biosynthetic demands of cancer cell proliferation. The proposed strategy may be implemented as a synergistic metabolic priming baseline in GBM as well as other tumors driven by glycolysis and glutaminolysis, regardless of their residual mitochondrial function. Suggested best practices are provided to guide future KMT research in metabolic oncology, offering a shared, evidence-driven framework for observational and interventional studies.
Collapse
Affiliation(s)
- Tomás Duraj
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.
| | | | - Giulio Zuccoli
- Neuroradiology, Private Practice, Philadelphia, PA, 19103, USA
| | - Joseph C Maroon
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Adrienne C Scheck
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Phoenix, AZ, 85004, USA
| | - Angela Poff
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Sebastian F Winter
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Jethro Hu
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, 97422, Schweinfurt, Germany
| | | | - Derek C Lee
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Isabella Cooper
- Ageing Biology and Age-Related Diseases Group, School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstr. 48, 5020, Salzburg, Austria
| | - Kenneth A Schwartz
- Department of Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Matthew C L Phillips
- Department of Neurology, Waikato Hospital, Hamilton, 3204, New Zealand
- Department of Medicine, University of Auckland, Auckland, 1142, New Zealand
| | - Colin E Champ
- Exercise Oncology & Resiliency Center and Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA, 15212, USA
| | | | - Jocelyn Tan-Shalaby
- School of Medicine, University of Pittsburgh, Veteran Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Fabiano M Serfaty
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, 20550-170, Brazil
- Serfaty Clínicas, Rio de Janeiro, RJ, 22440-040, Brazil
| | - Egiroh Omene
- Department of Oncology, Cross Cancer Institute, Edmonton, AB, T6G 1Z2, Canada
| | - Gabriel Arismendi-Morillo
- Department of Medicine, Faculty of Health Sciences, University of Deusto, 48007, Bilbao (Bizkaia), Spain
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, 4005, Venezuela
| | | | - Richard Cheng
- Cheng Integrative Health Center, Columbia, SC, 29212, USA
| | - Ahmed M El-Sakka
- Metabolic Terrain Institute of Health, East Congress Street, Tucson, AZ, 85701, USA
| | - Axel Pflueger
- Pflueger Medical Nephrologyand , Internal Medicine Services P.L.L.C, 6 Nelson Road, Monsey, NY, 10952, USA
| | - Edward H Mathews
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0002, South Africa
| | | | - Hanping Shi
- Department of Gastrointestinal Surgery and Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Raffaele Ivan Cincione
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Puglia, Italy
| | - Jean Pierre Spinosa
- Integrative Oncology, Breast and Gynecologic Oncology Surgery, Private Practice, Rue Des Terreaux 2, 1002, Lausanne, Switzerland
| | | | - Mehmet Salih Iyikesici
- Department of Medical Oncology, Altınbaş University Bahçelievler Medical Park Hospital, Istanbul, 34180, Turkey
| | - Atsuo Yanagisawa
- The Japanese College of Intravenous Therapy, Tokyo, 150-0013, Japan
| | | | - Anthony Chaffee
- Department of Neurosurgery, Sir Charles Gairdner Hospital, Perth, 6009, Australia
| | - Wafaa Abdel-Hadi
- Clinical Oncology Department, Cairo University, Giza, 12613, Egypt
| | - Amr K Elsamman
- Neurosurgery Department, Cairo University, Giza, 12613, Egypt
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, 6410 Rockledge Drive, Suite 610, Bethesda, MD, 20817, USA
| | - Keisuke Hagihara
- Department of Advanced Hybrid Medicine, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Zsófia Clemens
- International Center for Medical Nutritional Intervention, Budapest, 1137, Hungary
| | - George W Yu
- George W, Yu Foundation For Nutrition & Health and Aegis Medical & Research Associates, Annapolis, MD, 21401, USA
| | - Athanasios E Evangeliou
- Department of Pediatrics, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, Efkarpia, 56403, Thessaloniki, Greece
| | - Janak K Nathan
- Dr. DY Patil Medical College, Hospital and Research Centre, Pune, Maharashtra, 411018, India
| | - Kris Smith
- Barrow Neurological Institute, Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - David Fortin
- Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
| | - Jorg Dietrich
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | | | | |
Collapse
|
4
|
Mi X, Duan Y, Sun J, Tai Q, Yao H, Meng L, Yang X, Shi X, Shi B, Chen J, Sun L, Zhou D, Xiao S, Yao Y, He S. The ketogenic diet modulates tumor-associated neutrophil polarization via the AMOT-YAP/TAZ axis to inhibit colorectal cancer progression. Pharmacol Res 2024; 210:107494. [PMID: 39510146 DOI: 10.1016/j.phrs.2024.107494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Despite significant advances in the diagnosis and treatment of colorectal cancer (CRC), the prognosis for late-stage patients remains poor, highlighting the urgent need for new preventive and therapeutic strategies. Recent studies have focused on the ketogenic diet (KD) and its metabolite, β-hydroxybutyrate (BHB), for their tumor-suppressive effects and modulation of inflammatory responses. Using the azoxymethane (AOM) / dextran sulfate sodium (DSS)-induced mouse CRC model, we found that the ketogenic diet and BHB inhibit pro-tumor N2-type tumor-associated neutrophils (TANs) while promoting the polarization of TANs towards the anti-tumor N1 type. This shift in TANs polarization affects tumor growth and metastasis. The underlying mechanism involves BHB acting on the intracellular receptor histone deacetylases 3 (HDAC3), which modulates the activation of the AMOT-YAP/TAZ axis, leading to the inhibition of pro-carcinogenic factor transcription and release. Moreover, clinical cohort data corroborate these findings, showing that CRC patients with elevated BHB levels have significantly lower rates of lymph node involvement, which is associated with a higher infiltration ratio of anti-carcinogenic N1-type TANs in the tumor microenvironment (TME). These results suggest that BHB levels could serve as a prognostic biomarker for CRC. In conclusion, our findings indicate that BHB derived from KD regulates TANs polarization in CRC via the HDAC3-AMOT-YAP/TAZ axis, effectively inhibiting tumor growth and metastasis. These insights establish a novel theoretical basis for employing the KD in the treatment of CRC and for developing cancer adjuvant immunotherapy strategy based on the polarization of neutrophils.
Collapse
Affiliation(s)
- Xiuwei Mi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Yudong Duan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiying Sun
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China; Department of Respiratory Diseases, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, China
| | - Qingliang Tai
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Lijun Meng
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Xiaoshan Yang
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Xinyu Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Bo Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Junjie Chen
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215299, China
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Diyuan Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
5
|
Deng Q, Lv R, Zou T. The effects of the ketogenic diet on cancer treatment: a narrative review. Eur J Cancer Prev 2024:00008469-990000000-00175. [PMID: 39365252 DOI: 10.1097/cej.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Despite significant advances in therapy, cancer remains the top cause of death in parts of the globe. For many types of cancer, the typical treatment is a combination of surgery, chemotherapy, and radiotherapy. However, this conventional treatment is not successful on its own. As a consequence, innovative approaches that improve treatment efficacy are urgently needed. The ketogenic diet is a high-fat, moderate protein, and low-carbohydrate diet that appears to sensitize most cancers to conventional therapies by exploiting cancer cells' altered metabolism, making it an effective adjuvant cancer treatment alternative. This diet could decrease glucose metabolism while enhancing lipid metabolism, interfering with the Warburg effect, and inhibiting tumor cell proliferation. The anticancer impact of ketogenic diet has been established in numerous animal trials and clinical investigations on a wide range of tumor types, including glioblastoma, pancreatic cancer, head and neck cancer, breast cancer, invasive rectal cancer, ovarian cancer, and endometrial cancer. In this review, we discussed the various types of ketogenic diets, the mechanism of action for ketogenic diet as a cancer therapy, and the data gathered from continuing preclinical and clinical studies, intending to establish a solid theoretical foundation for future research.
Collapse
Affiliation(s)
- Qingxuan Deng
- Dongguan Key Laboratory of Environmental Medicine, Institute of Medical Systems Biology, School of Public Health
| | - Ruyue Lv
- Dongguan Key Laboratory of Environmental Medicine, Institute of Medical Systems Biology, School of Public Health
| | - Tangbin Zou
- Dongguan Key Laboratory of Environmental Medicine, Institute of Medical Systems Biology, School of Public Health
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
6
|
Valerio J, Borro M, Proietti E, Pisciotta L, Olarinde IO, Fernandez Gomez M, Alvarez Pinzon AM. Systematic Review and Clinical Insights: The Role of the Ketogenic Diet in Managing Glioblastoma in Cancer Neuroscience. J Pers Med 2024; 14:929. [PMID: 39338183 PMCID: PMC11433106 DOI: 10.3390/jpm14090929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/02/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
Recent scientific research has shown that the ketogenic diet may have potential benefits in a variety of medical fields, which has led to the diet receiving a substantial amount of attention. Clinical and experimental research on brain tumors has shown that the ketogenic diet has a satisfactory safety profile. This safety profile has been established in a variety of applications, including the management of obesity and the treatment of drug-resistant epileptic cases. However, in human studies, the impact of ketogenic therapy on the growth of tumors and the life expectancy of patients has not provided results that are well characterized. Consequently, our purpose is to improve the comprehension of these features by succinctly presenting the developments and conclusions that have been gained from the most recent study that pertains to this non-pharmacological technique. According to the findings of our study, patients with brain tumors who stick to a ketogenic diet are more likely to experience improved survival rates. However, it is required to conduct additional research on humans in order to more accurately define the anti-tumor efficiency of this diet as well as the underlying processes that support the therapeutic effects of this dieting regimen.
Collapse
Affiliation(s)
- Jose Valerio
- Neurosurgery Oncology Center of Excellence, Neurosurgery Department, Miami Neuroscience Center at Larkin, South Miami, FL 33143, USA
| | - Matteo Borro
- Internal Medicine Unit, Department of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Elisa Proietti
- Department of Internal Medicine (DIMI), University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | - Livia Pisciotta
- Department of Internal Medicine (DIMI), University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- Operative Unit of Dietetics and Clinical Nutrition, Department of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Immanuel O Olarinde
- Neurosurgery Department, Latino America Valerio Foundation, Weston, FL 33331, USA
| | | | - Andres Mauricio Alvarez Pinzon
- MCIFAU Cancer Center of Excellence, Memorial Cancer Institute, Memorial Healthcare System, Hollywood, FL 33021, USA
- Cancer Neuroscience Program, The Institute of Neuroscience of Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute for Human Health and Disease Intervention, Division of Research, FAU Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
7
|
Radulescu D, Mihai FD, Trasca MET, Caluianu EI, Calafeteanu CDM, Radulescu PM, Mercut R, Ciupeanu-Calugaru ED, Marinescu GA, Siloşi CA, Nistor CCE, Danoiu S. Oxidative Stress in Military Missions-Impact and Management Strategies: A Narrative Analysis. Life (Basel) 2024; 14:567. [PMID: 38792589 PMCID: PMC11121804 DOI: 10.3390/life14050567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
This narrative review comprehensively examines the impact of oxidative stress on military personnel, highlighting the crucial role of physical exercise and tailored diets, particularly the ketogenic diet, in minimizing this stress. Through a meticulous analysis of the recent literature, the study emphasizes how regular physical exercise not only enhances cardiovascular, cognitive, and musculoskeletal health but is also essential in neutralizing the effects of oxidative stress, thereby improving endurance and performance during long-term missions. Furthermore, the implementation of the ketogenic diet provides an efficient and consistent energy source through ketone bodies, tailored to the specific energy requirements of military activities, and significantly contributes to the reduction in reactive oxygen species production, thus protecting against cellular deterioration under extreme stress. The study also underlines the importance of integrating advanced technologies, such as wearable devices and smart sensors that allow for the precise and real-time monitoring of oxidative stress and physiological responses, thus facilitating the customization of training and nutritional regimes. Observations from this review emphasize significant variability among individuals in responses to oxidative stress, highlighting the need for a personalized approach in formulating intervention strategies. It is crucial to develop and implement well-monitored, personalized supplementation protocols to ensure that each member of the military personnel receives a regimen tailored to their specific needs, thereby maximizing the effectiveness of measures to combat oxidative stress. This analysis makes a valuable contribution to the specialized literature, proposing a detailed framework for addressing oxidative stress in the armed forces and opening new directions for future research with the aim of optimizing clinical practices and improving the health and performance of military personnel under stress and specific challenges of the military field.
Collapse
Affiliation(s)
- Dumitru Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Florina-Diana Mihai
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Major Emil-Tiberius Trasca
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Elena-Irina Caluianu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Captain Dan Marian Calafeteanu
- Department of Ortopedics, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania;
| | - Patricia-Mihaela Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Razvan Mercut
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | - Georgiana-Andreea Marinescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Cristian-Adrian Siloşi
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | | | - Suzana Danoiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
8
|
Guo L, Zhang B, Zhang W, Xie Y, Chen X, Sun X, Watt DS, Liu C, Spielmann HP, Liu X. Inhibition of Carbohydrate Metabolism Potentiated by the Therapeutic Effects of Oxidative Phosphorylation Inhibitors in Colon Cancer Cells. Cancers (Basel) 2024; 16:1399. [PMID: 38611076 PMCID: PMC11010912 DOI: 10.3390/cancers16071399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer cells undergo a significant level of "metabolic reprogramming" or "remodeling" to ensure an adequate supply of ATP and "building blocks" for cell survival and to facilitate accelerated proliferation. Cancer cells preferentially use glycolysis for ATP production (the Warburg effect); however, cancer cells, including colorectal cancer (CRC) cells, also depend on oxidative phosphorylation (OXPHOS) for ATP production, a finding that suggests that both glycolysis and OXPHOS play significant roles in facilitating cancer progression and proliferation. Our prior studies identified a semisynthetic isoflavonoid, DBI-1, that served as an AMPK activator targeting mitochondrial complex I. Furthermore, DBI-1 and a glucose transporter 1 (GLUT1) inhibitor, BAY-876, synergistically inhibited CRC cell growth in vitro and in vivo. We now report a study of the structure-activity relationships (SARs) in the isoflavonoid family in which we identified a new DBI-1 analog, namely, DBI-2, with promising properties. Here, we aimed to explore the antitumor mechanisms of DBIs and to develop new combination strategies by targeting both glycolysis and OXPHOS. We identified DBI-2 as a novel AMPK activator using an AMPK phosphorylation assay as a readout. DBI-2 inhibited mitochondrial complex I in the Seahorse assays. We performed proliferation and Western blotting assays and conducted studies of apoptosis, necrosis, and autophagy to corroborate the synergistic effects of DBI-2 and BAY-876 on CRC cells in vitro. We hypothesized that restricting the carbohydrate uptake with a KD would mimic the effects of GLUT1 inhibitors, and we found that a ketogenic diet significantly enhanced the therapeutic efficacy of DBI-2 in CRC xenograft mouse models, an outcome that suggested a potentially new approach for combination cancer therapy.
Collapse
Affiliation(s)
- Lichao Guo
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory and Center for Drug Innovation and Discovery, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Baochen Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory and Center for Drug Innovation and Discovery, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Wen Zhang
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Yanqi Xie
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Xi Chen
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory and Center for Drug Innovation and Discovery, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Xueke Sun
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory and Center for Drug Innovation and Discovery, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - David S. Watt
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Chunming Liu
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - H. Peter Spielmann
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Xifu Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory and Center for Drug Innovation and Discovery, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| |
Collapse
|
9
|
Klement RJ. Anti-tumor effects of ketogenic diets and their synergism with other treatments in mice: Bayesian evidence synthesis of 1755 individual mouse survival data. Biomed J 2024; 47:100609. [PMID: 37245566 PMCID: PMC10900256 DOI: 10.1016/j.bj.2023.100609] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Ketogenic diets (KDs) are high-fat diets with putative anti-tumor effects. The aim of this study was to synthesize the evidence for the anti-tumor effects of KDs in mice, with a focus on their possible synergism with chemotherapy (CT), radiotherapy (RT), or targeted therapies (TT). METHODS Relevant studies were retrieved from a literature search. A total of 43 articles reporting on 65 mouse experiments fulfilled the inclusion criteria, and 1755 individual mouse survival times were collated from the study authors or the publications. The restricted mean survival time ratio (RMSTR) between the KD and control groups served as the effect size. Bayesian evidence synthesis models were used to estimate pooled effect sizes and to assess the impact of putative confounders and synergism between KD and other therapies. RESULTS Overall, there was a significant survival-prolonging effect of KD monotherapy (RMSTR = 1.161 ± 0.040), which was confirmed in meta-regression accounting for syngeneic versus xenogeneic models, early versus late KD start and subcutaneous versus other organ growth. Combining the KD with RT or TT, but not CT, was associated with a further 30% (RT) or 21% (TT) prolongation of survival. An analysis accounting for 15 individual tumor entities showed that KDs exerted significant survival-prolonging effects in pancreatic cancer (all treatment combinations), gliomas (KD + RT and KD + TT), head and neck cancer (KD + RT), and stomach cancer (KD+RT and KD + TT). CONCLUSIONS This analytical study confirmed the overall anti-tumor effects of KDs in a large number of mouse experiments and provides evidence for synergistic effects with RT and TT.
Collapse
Affiliation(s)
- Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Schweinfurt, Germany.
| |
Collapse
|
10
|
Filippou C, Themistocleous SC, Marangos G, Panayiotou Y, Fyrilla M, Kousparou CA, Pana ZD, Tsioutis C, Johnson EO, Yiallouris A. Microbial Therapy and Breast Cancer Management: Exploring Mechanisms, Clinical Efficacy, and Integration within the One Health Approach. Int J Mol Sci 2024; 25:1110. [PMID: 38256183 PMCID: PMC10816061 DOI: 10.3390/ijms25021110] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
This comprehensive review elucidates the profound relationship between the human microbiome and breast cancer management. Recent findings highlight the significance of microbial alterations in tissue, such as the gut and the breast, and their role in influencing the breast cancer risk, development, progression, and treatment outcomes. We delve into how the gut microbiome can modulate systemic inflammatory responses and estrogen levels, thereby impacting cancer initiation and therapeutic drug efficacy. Furthermore, we explore the unique microbial diversity within breast tissue, indicating potential imbalances brought about by cancer and highlighting specific microbes as promising therapeutic targets. Emphasizing a holistic One Health approach, this review underscores the importance of integrating insights from human, animal, and environmental health to gain a deeper understanding of the complex microbe-cancer interplay. As the field advances, the strategic manipulation of the microbiome and its metabolites presents innovative prospects for the enhancement of cancer diagnostics and therapeutics. However, rigorous clinical trials remain essential to confirm the potential of microbiota-based interventions in breast cancer management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Andreas Yiallouris
- School of Medicine, European University Cyprus, 6 Diogenis Str., 2404 Engomi, P.O. Box 22006, Nicosia 1516, Cyprus
| |
Collapse
|
11
|
Buga A, Harper DG, Sapper TN, Hyde PN, Fell B, Dickerson R, Stoner JT, Kackley ML, Crabtree CD, Decker DD, Robinson BT, Krystal G, Binzel K, Lustberg MB, Volek JS. Feasibility and metabolic outcomes of a well-formulated ketogenic diet as an adjuvant therapeutic intervention for women with stage IV metastatic breast cancer: The Keto-CARE trial. PLoS One 2024; 19:e0296523. [PMID: 38166036 PMCID: PMC10760925 DOI: 10.1371/journal.pone.0296523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/13/2023] [Indexed: 01/04/2024] Open
Abstract
PURPOSE Ketogenic diets may positively influence cancer through pleiotropic mechanisms, but only a few small and short-term studies have addressed feasibility and efficacy in cancer patients. The primary goals of this study were to evaluate the feasibility and the sustained metabolic effects of a personalized well-formulated ketogenic diet (WFKD) designed to achieve consistent blood beta-hydroxybutyrate (βHB) >0.5 mM in women diagnosed with stage IV metastatic breast cancer (MBC) undergoing chemotherapy. METHODS Women (n = 20) were enrolled in a six month, two-phase, single-arm WFKD intervention (NCT03535701). Phase I was a highly-supervised, ad libitum, personalized WFKD, where women were provided with ketogenic-appropriate food daily for three months. Phase II transitioned women to a self-administered WFKD with ongoing coaching for an additional three months. Fasting capillary βHB and glucose were collected daily; weight, body composition, plasma insulin, and insulin resistance were collected at baseline, three and six months. RESULTS Capillary βHB indicated women achieved nutritional ketosis (Phase I mean: 0.8 mM (n = 15); Phase II mean: 0.7 mM (n = 9)). Body weight decreased 10% after three months, primarily from body fat. Fasting plasma glucose, plasma insulin, and insulin resistance also decreased significantly after three months (p < 0.01), an effect that persisted at six months. CONCLUSIONS Women diagnosed with MBC undergoing chemotherapy can safely achieve and maintain nutritional ketosis, while improving body composition and insulin resistance, out to six months.
Collapse
Affiliation(s)
- Alex Buga
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - David G. Harper
- School of Kinesiology, University of the Fraser Valley, Abbotsford, British Columbia, Canada
| | - Teryn N. Sapper
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Parker N. Hyde
- Department of Kinesiology, University of North Georgia, Dahlonega, Georgia, United States of America
| | - Brandon Fell
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Ryan Dickerson
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Justen T. Stoner
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Madison L. Kackley
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Christopher D. Crabtree
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Drew D. Decker
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Bradley T. Robinson
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Gerald Krystal
- The Terry Fox Laboratory, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Katherine Binzel
- Department of Radiology, Wright Center of Innovation, The Ohio State University, Columbus, Ohio, United States of America
| | - Maryam B. Lustberg
- Breast Cancer Center, Smilow Cancer Hospital, Yale University, New Haven, Connecticut, United States of America
| | - Jeff S. Volek
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
12
|
Shahtaghi NR, Soni B, Bakrey H, Bigdelitabar S, Jain SK. Beta-Hydroxybutyrate: A Supplemental Molecule for Various Diseases. Curr Drug Targets 2024; 25:919-933. [PMID: 39238395 DOI: 10.2174/0113894501312168240821082224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/04/2024] [Accepted: 07/25/2024] [Indexed: 09/07/2024]
Abstract
β-hydroxybutyrate (BHB) is a ketone body that serves as an alternative energy source for various tissues, including the brain, heart, and skeletal muscle. As a metabolic intermediate and signaling molecule, BHB plays a crucial role in modulating cellular and physiological processes. Notably, BHB supplementation offers a novel and promising strategy to induce nutritional ketosis without the need for strict dietary adherence or causing nutritional deficiencies. This review article provides an overview of BHB metabolism and explores its applications in age-related diseases. This review conducted a comprehensive search of PubMed, ScienceDirect, and other relevant English-language articles. The main findings were synthesized, and discussed the challenges, limitations, and future directions of BHB supplementation. BHB supplementation holds potential benefits for various diseases and conditions, including neurodegenerative disorders, cardiovascular diseases, cancers, and inflammation. BHB acts through multiple mechanisms, including interactions with cell surface receptors, intracellular enzymes, transcription factors, signaling molecules, and epigenetic modifications. Despite its promise, BHB supplementation faces several challenges, such as determining the optimal dosage, ensuring long-term safety, identifying the most effective type and formulation, establishing biomarkers of response, and conducting cost-effectiveness analyses. BHB supplementation opens exciting avenues for research, including investigating molecular mechanisms, refining optimization strategies, exploring innovation opportunities, and assessing healthspan and lifespan benefits. BHB supplementation represents a new frontier in health research, offering a potential pathway to enhance well-being and extend lifespan.
Collapse
Affiliation(s)
- Navid Reza Shahtaghi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, 143005, Amritsar, Punjab, India
| | - Bindu Soni
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, 143005, Amritsar, Punjab, India
| | - Hossamaldeen Bakrey
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, 143005, Amritsar, Punjab, India
| | - Samira Bigdelitabar
- Department of Microbiology, Government Medical College, 143001, Amritsar, Punjab, India
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, 143005, Amritsar, Punjab, India
- Centre for Basic & Translational Research in Health Sciences (CBTHRS), Guru Nanak Dev University, 143005, Amritsar, Punjab, India
| |
Collapse
|
13
|
Zhou X, Wang Z, Yuan K. The effect of diet and nutrition on T cell function in cancer. Int J Cancer 2023; 153:1954-1966. [PMID: 37504380 DOI: 10.1002/ijc.34668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023]
Abstract
Cancer can be considered one of the most threatening diseases to human health, and immunotherapy, especially T-cell immunotherapy, is the most promising treatment for cancers. Diet therapy is widely concerned in cancer because of its safety and fewer side effects. Many studies have shown that both the function of T cells and the progression of cancer can be affected by nutrients in the diet. In fact, it is challenging for T cells to infiltrate and eliminate cancer cells in tumor microenvironment, because of the harsh metabolic condition. The intake of different nutrients has a great influence on the proliferation, activation, differentiation and exhaustion of T cells. In this review, we summarize the effects of typical amino acids, lipids, carbohydrates and other nutritional factors on T cell functions and provide future perspectives for dietary treatment of cancer based on modifications of T cell functions.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Wang
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Halma MTJ, Tuszynski JA, Marik PE. Cancer Metabolism as a Therapeutic Target and Review of Interventions. Nutrients 2023; 15:4245. [PMID: 37836529 PMCID: PMC10574675 DOI: 10.3390/nu15194245] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Cancer is amenable to low-cost treatments, given that it has a significant metabolic component, which can be affected through diet and lifestyle change at minimal cost. The Warburg hypothesis states that cancer cells have an altered cell metabolism towards anaerobic glycolysis. Given this metabolic reprogramming in cancer cells, it is possible to target cancers metabolically by depriving them of glucose. In addition to dietary and lifestyle modifications which work on tumors metabolically, there are a panoply of nutritional supplements and repurposed drugs associated with cancer prevention and better treatment outcomes. These interventions and their evidentiary basis are covered in the latter half of this review to guide future cancer treatment.
Collapse
Affiliation(s)
- Matthew T. J. Halma
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- EbMC Squared CIC, Bath BA2 4BL, UK
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, I-1029 Turin, Italy
| | - Paul E. Marik
- Frontline COVID-19 Critical Care Alliance, Washington, DC 20036, USA
| |
Collapse
|
15
|
Bezerra LS, Santos-Veloso MAO. Ketogenic diet and metastasis: A critical review of the literature and possible mechanisms. Clin Nutr ESPEN 2023; 57:207-212. [PMID: 37739657 DOI: 10.1016/j.clnesp.2023.06.038] [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: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 09/24/2023]
Abstract
The ketogenic diet (KD) is a low-carb diet that has been indicated as a possible coadjuvant in cancer therapy, mainly due to its capacity to reduce glycolysis production, inflammation, and oxidative stress. However, KD's role in metastasis remains poorly explored. This study aims to provide a critical review of the literature about KD's efficacy in metastasis therapy and the possible molecular mechanisms behind it. Initially, general concepts on KD and metastasis are discussed. Then, it delves deeper into the main cancer mechanisms explored by KD experimental studies, discussing the central results obtained in metastasis research and their main limiting conditions. Following, there is a critical analysis of clinical trials, including those in the grey literature. In the end, there is a summary of the actual studies' limitations and barriers to future research. To date, it is possible to conclude that there is not enough evidence supporting the efficacy of KD in the treatment of metastasis.
Collapse
Affiliation(s)
- Lucas Soares Bezerra
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Postgraduate Program in Therapeutic Innovation, Biosciences Center, Federal University of Pernambuco, Recife, Brazil.
| | | |
Collapse
|
16
|
Alpuim Costa D, Gonçalves-Nobre JG, Sampaio-Alves M, Guerra N, Arana Ribeiro J, Espiney Amaro C. Hyperbaric oxygen therapy as a complementary treatment in neuroblastoma - a narrative review. Front Oncol 2023; 13:1254322. [PMID: 37823059 PMCID: PMC10562625 DOI: 10.3389/fonc.2023.1254322] [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: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Neuroblastoma is the most frequently diagnosed cancer during the first year of life. This neoplasm originates from neural crest cells derived from the sympathetic nervous system, adrenal medulla, or paraspinal ganglia. The clinical presentation can vary from an asymptomatic mass to symptoms resulting from local invasion and/or spread of distant disease spread. The natural history of neuroblastoma is highly variable, ranging from relatively indolent biological behavior to a high-risk clinical phenotype with a dismal prognosis. Age, stage, and biological features are important prognostic risk stratification and treatment assignment prognostic factors. The multimodal therapy approach includes myeloablative chemotherapy, radiotherapy, immunotherapy, and aggressive surgical resection. Hyperbaric oxygen therapy (HBOT) has been proposed as a complementary measure to overcome tumor hypoxia, which is considered one of the hallmarks of this cancer treatment resistance. This article aims to review the relevant literature on the neuroblastoma pathophysiology, clinical presentation, and different biological and genetic profiles, and to discuss its management, focusing on HBOT.
Collapse
Affiliation(s)
- Diogo Alpuim Costa
- Hematology and Oncology Department, CUF Oncologia, Lisbon, Portugal
- Centro de Medicina Subaquática e Hiperbárica (CMSH), Portuguese Navy, Lisbon, Portugal
- Medical Oncology Department, Hospital de Cascais Dr. José de Almeida, Alcabideche, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade NOVA de Lisboa, Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - J. Guilherme Gonçalves-Nobre
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Hospital Garcia de Orta (HGO), E.P.E., Almada, Portugal
- Instituto de Saúde Ambiental (ISAMB), Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- Instituto de Medicina Preventiva & Saúde Pública (IMP&SP), Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- PTSurg – Portuguese Surgical Research Collaborative, Lisboa, Portugal PTSurg – Portuguese Surgical Research Collaborative, Lisbon, Portugal
| | - Mafalda Sampaio-Alves
- PTSurg – Portuguese Surgical Research Collaborative, Lisboa, Portugal PTSurg – Portuguese Surgical Research Collaborative, Lisbon, Portugal
- Faculty of Medicine, University of Porto, Oporto, Portugal
| | - Nuno Guerra
- Centro de Medicina Subaquática e Hiperbárica (CMSH), Portuguese Navy, Lisbon, Portugal
| | | | - Carla Espiney Amaro
- Centro de Medicina Subaquática e Hiperbárica (CMSH), Portuguese Navy, Lisbon, Portugal
| |
Collapse
|
17
|
Directo D, Lee SR. Cancer Cachexia: Underlying Mechanisms and Potential Therapeutic Interventions. Metabolites 2023; 13:1024. [PMID: 37755304 PMCID: PMC10538050 DOI: 10.3390/metabo13091024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer cachexia, a multifactorial metabolic syndrome developed during malignant tumor growth, is characterized by an accelerated loss of body weight accompanied by the depletion of skeletal muscle mass. This debilitating condition is associated with muscle degradation, impaired immune function, reduced functional capacity, compromised quality of life, and diminished survival in cancer patients. Despite the lack of the known capability of fully reversing or ameliorating this condition, ongoing research is shedding light on promising preclinical approaches that target the disrupted mechanisms in the pathophysiology of cancer cachexia. This comprehensive review delves into critical aspects of cancer cachexia, including its underlying pathophysiological mechanisms, preclinical models for studying the progression of cancer cachexia, methods for clinical assessment, relevant biomarkers, and potential therapeutic strategies. These discussions collectively aim to contribute to the evolving foundation for effective, multifaceted counteractive strategies against this challenging condition.
Collapse
Affiliation(s)
| | - Sang-Rok Lee
- Department of Kinesiology, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
18
|
Miller AI, Diaz D, Lin B, Krzesaj PK, Ustoyev S, Shim A, Fine EJ, Sarafraz-Yazdi E, Pincus MR, Feinman RD. Ketone Bodies Induce Unique Inhibition of Tumor Cell Proliferation and Enhance the Efficacy of Anti-Cancer Agents. Biomedicines 2023; 11:2515. [PMID: 37760956 PMCID: PMC10526402 DOI: 10.3390/biomedicines11092515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The ketone bodies, sodium and lithium salts of acetoacetate (AcAc) and sodium 3-hydroxybutyrate (3-HB; commonly called beta-hydroxybutyrate) have been found to inhibit the proliferation of cancer cells. Previous studies have suggested that lithium itself may be an inhibiting agent but may be additive or synergistic with the effect of AcAc. We previously found that sodium acetoacetate (NaAcAc) inhibits the growth of human colon cancer cell line SW480. We report here similar results for several other cancer cell lines including ovarian, cervical and breast cancers. We found that NaAcAc does not kill cancer cells but rather blocks their proliferation. Similar inhibition of growth was seen in the effect of lithium ion alone (as LiCl). The effect of LiAcAc appears to be due to the combined effects of acetoacetate and the lithium ion. The ketone bodies, when given together with chemotherapeutic agents, rapamycin, methotrexate and the new peptide anti-cancer agent, PNC-27, substantially lowers their IC50 values for cancer cell, killing suggesting that ketone bodies and ketogenic diets may be powerful adjunct agents in treating human cancers.
Collapse
Affiliation(s)
- Anna I. Miller
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - David Diaz
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Bo Lin
- Department of Pathology, AdventHealth, 301 Memorial Medical Pkwy, Daytona Beach, FL 32117, USA
| | - Patryk K. Krzesaj
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Sarah Ustoyev
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Alfred Shim
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Eugene J. Fine
- Department of Radiology (Nuclear Medicine), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ehsan Sarafraz-Yazdi
- NomoCan Pharmaceuticals LLC, New York Blood Center, 310 East 67th Street, New York, NY 10065, USA
| | - Matthew R. Pincus
- Department of Pathology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Richard D. Feinman
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| |
Collapse
|
19
|
Vinkel J, Arenkiel B, Hyldegaard O. The Mechanisms of Action of Hyperbaric Oxygen in Restoring Host Homeostasis during Sepsis. Biomolecules 2023; 13:1228. [PMID: 37627293 PMCID: PMC10452474 DOI: 10.3390/biom13081228] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The perception of sepsis has shifted over time; however, it remains a leading cause of death worldwide. Sepsis is now recognized as an imbalance in host cellular functions triggered by the invading pathogens, both related to immune cells, endothelial function, glucose and oxygen metabolism, tissue repair and restoration. Many of these key mechanisms in sepsis are also targets of hyperbaric oxygen (HBO2) treatment. HBO2 treatment has been shown to improve survival in clinical studies on patients with necrotizing soft tissue infections as well as experimental sepsis models. High tissue oxygen tension during HBO2 treatment may affect oxidative phosphorylation in mitochondria. Oxygen is converted to energy, and, as a natural byproduct, reactive oxygen species are produced. Reactive oxygen species can act as mediators, and both these and the HBO2-mediated increase in oxygen supply have the potential to influence the cellular processes involved in sepsis. The pathophysiology of sepsis can be explained comprehensively through resistance and tolerance to infection. We argue that HBO2 treatment may protect the host from collateral tissue damage during resistance by reducing neutrophil extracellular traps, inhibiting neutrophil adhesion to vascular endothelium, reducing proinflammatory cytokines, and halting the Warburg effect, while also assisting the host in tolerance to infection by reducing iron-mediated injury and upregulating anti-inflammatory measures. Finally, we show how inflammation and oxygen-sensing pathways are connected on the cellular level in a self-reinforcing and detrimental manner in inflammatory conditions, and with support from a substantial body of studies from the literature, we conclude by demonstrating that HBO2 treatment can intervene to maintain homeostasis.
Collapse
Affiliation(s)
- Julie Vinkel
- Department of Anesthesiology, Centre of Head and Orthopedics, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bjoern Arenkiel
- Department of Anesthesiology, Centre of Head and Orthopedics, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Ole Hyldegaard
- Department of Anesthesiology, Centre of Head and Orthopedics, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
20
|
Cortez NE, Pathak S, Rodriguez Lanzi C, Hong BV, Crone R, Sule R, Wang F, Chen S, Gomes AV, Baar K, Mackenzie GG. A Ketogenic Diet in Combination with Gemcitabine Mitigates Pancreatic Cancer-Associated Cachexia in Male and Female KPC Mice. Int J Mol Sci 2023; 24:10753. [PMID: 37445930 PMCID: PMC10341838 DOI: 10.3390/ijms241310753] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a critical contributor to pancreatic ductal adenocarcinoma (PDAC) mortality. Thus, there is an urgent need for new strategies to mitigate PDAC-associated cachexia; and the exploration of dietary interventions is a critical component. We previously observed that a ketogenic diet (KD) combined with gemcitabine enhances overall survival in the autochthonous LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx1-Cre (KPC) mouse model. In this study, we investigated the effect and cellular mechanisms of a KD in combination with gemcitabine on the maintenance of skeletal muscle mass in KPC mice. For this purpose, male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD), a KD, a CD + gemcitabine (CG), or a KD + gemcitabine (KG) group. We observed that a KD or a KG-mitigated muscle strength declined over time and presented higher gastrocnemius weights compared CD-fed mice. Mechanistically, we observed sex-dependent effects of KG treatment, including the inhibition of autophagy, and increased phosphorylation levels of eIF2α in KG-treated KPC mice when compared to CG-treated mice. Our data suggest that a KG results in preservation of skeletal muscle mass. Additional research is warranted to explore whether this diet-treatment combination can be clinically effective in combating CAC in PDAC patients.
Collapse
Affiliation(s)
- Natalia E. Cortez
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
| | - Suraj Pathak
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Cecilia Rodriguez Lanzi
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
| | - Brian V. Hong
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
| | - Ryman Crone
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Rasheed Sule
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Fangyi Wang
- Department of Animal Science, University of California, One Shields Ave., Davis, CA 95616, USA;
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, University of California, One Shields Ave., Davis, CA 95616, USA;
- University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Aldrin V. Gomes
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Keith Baar
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Gerardo G. Mackenzie
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
- University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| |
Collapse
|
21
|
Al-Jada DN, Takruri HR, Talib WH. From antiepileptic therapy to promising adjuvant in medical oncology: A historical view of the ketogenic diet. PHARMANUTRITION 2023. [DOI: 10.1016/j.phanu.2023.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
22
|
Nadhan R, Kashyap S, Ha JH, Jayaraman M, Song YS, Isidoro C, Dhanasekaran DN. Targeting Oncometabolites in Peritoneal Cancers: Preclinical Insights and Therapeutic Strategies. Metabolites 2023; 13:618. [PMID: 37233659 PMCID: PMC10222714 DOI: 10.3390/metabo13050618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Peritoneal cancers present significant clinical challenges with poor prognosis. Understanding the role of cancer cell metabolism and cancer-promoting metabolites in peritoneal cancers can provide new insights into the mechanisms that drive tumor progression and can identify novel therapeutic targets and biomarkers for early detection, prognosis, and treatment response. Cancer cells dynamically reprogram their metabolism to facilitate tumor growth and overcome metabolic stress, with cancer-promoting metabolites such as kynurenines, lactate, and sphingosine-1-phosphate promoting cell proliferation, angiogenesis, and immune evasion. Targeting cancer-promoting metabolites could also lead to the development of effective combinatorial and adjuvant therapies involving metabolic inhibitors for the treatment of peritoneal cancers. With the observed metabolomic heterogeneity in cancer patients, defining peritoneal cancer metabolome and cancer-promoting metabolites holds great promise for improving outcomes for patients with peritoneal tumors and advancing the field of precision cancer medicine. This review provides an overview of the metabolic signatures of peritoneal cancer cells, explores the role of cancer-promoting metabolites as potential therapeutic targets, and discusses the implications for advancing precision cancer medicine in peritoneal cancers.
Collapse
Affiliation(s)
- Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
| | - Srishti Kashyap
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
| | - Ji Hee Ha
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Muralidharan Jayaraman
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul 151-921, Republic of Korea
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
23
|
Ketogenic Diet Applied in Weight Reduction of Overweight and Obese Individuals with Progress Prediction by Use of the Modified Wishnofsky Equation. Nutrients 2023; 15:nu15040927. [PMID: 36839285 PMCID: PMC9968058 DOI: 10.3390/nu15040927] [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: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Ketogenic diet is often used as diet therapy for certain diseases, among other things, its positive effect related to weight loss is highlighted. Precisely because of the suggestion that KD can help with weight loss, visceral obesity, and appetite control, 100 respondents joined the weight loss program (of which 31% were men and 69% were women). The aforementioned respondents were interviewed in order to determine their eating habits, the amount of food consumed, and the time when they consume meals. Basic anthropometric data (body height, body mass, chest, waist, hips, biceps, and thigh circumferences) were also collected, in order to be able to monitor their progress during the different phases of the ketogenic diet. Important information is the expected body mass during the time frame of a certain keto diet phase. This information is important for the nutritionist, medical doctor, as well as for the participant in the reduced diet program; therefore, the model was developed that modified the original equation according to Wishnofsky. The results show that women lost an average of 22.7 kg (average number of days in the program 79.5), and for men the average weight loss was slightly higher, 29.7 kg (with an average of 76.8 days in the program). The prediction of expected body mass by the modified Wishnofsky's equation was extremely well aligned with the experimental values, as shown by the Bland-Altman graph (bias for women 0.021 kg and -0.697 kg for men) and the coefficient of determination of 0.9903. The modification of the Wishnofsky equation further shed light on the importance of controlled energy reduction during the dietetic options of the ketogenic diet.
Collapse
|
24
|
Tamraz M, Al Ghossaini N, Temraz S. The Ketogenic Diet in Colorectal Cancer: A Means to an End. Int J Mol Sci 2023; 24:ijms24043683. [PMID: 36835094 PMCID: PMC9965563 DOI: 10.3390/ijms24043683] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Some diets, such as high lipid and high glucose diets, are known to increase the risk of colorectal cancer. On the other hand, little is known about diets that prevent colonic carcinogenesis. The ketogenic diet, which is characterized by high fat and very low carbohydrate content, is one such diet. The ketogenic diet decreases the amount of available glucose for tumors and shifts to the production of ketone bodies as an alternative energy source for healthy cells. Cancer cells are unable to use the ketone bodies for energy thus depriving them of the energy needed for progression and survival. Many studies reported the beneficial effects of the ketogenic diet in several types of cancers. Recently, the ketone body β-hydroxybutyrate has been found to possess anti-tumor potential in colorectal cancer. Despite its beneficial effects, the ketogenic diet also has some drawbacks, some of which are related to gastrointestinal disorders and weight loss. Thus, studies are being directed at this time towards finding alternatives to following a strict ketogenic diet and supplementing patients with the ketone bodies responsible for its beneficial effects in the hope of overcoming some potential setbacks. This article discusses the mechanism by which a ketogenic diet influences growth and proliferation of tumor cells, it sheds the light on the most recent trials regarding its use as an adjunctive measure to chemotherapy in patients with metastatic colorectal cancer, and it explains the limitations of its usage in metastatic patients and the promising role of exogenous ketone supplementation in this setting.
Collapse
Affiliation(s)
- Magie Tamraz
- Department of Nutrition and Dietetics, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon
| | - Najib Al Ghossaini
- Department of Internal Medicine, Ain Wazein Medical Village, Chouf 5841, Lebanon
| | - Sally Temraz
- Department of Internal Medicine, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon
- Correspondence: ; Tel.: +961-1-374374
| |
Collapse
|
25
|
Makuku R, Sinaei Far Z, Khalili N, Moyo A, Razi S, Keshavarz-Fathi M, Mahmoudi M, Rezaei N. The Role of Ketogenic Diet in the Treatment of Neuroblastoma. Integr Cancer Ther 2023; 22:15347354221150787. [PMID: 36752115 PMCID: PMC9909060 DOI: 10.1177/15347354221150787] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/15/2022] [Indexed: 02/09/2023] Open
Abstract
The ketogenic diet (KD) was initially used in 1920 for drug-resistant epileptic patients. From this point onward, ketogenic diets became a pivotal part of nutritional therapy research. To date, KD has shown therapeutic potential in many pathologies such as Alzheimer's disease, Parkinson's disease, autism, brain cancers, and multiple sclerosis. Although KD is now an adjuvant therapy for certain diseases, its effectiveness as an antitumor nutritional therapy is still an ongoing debate, especially in Neuroblastoma. Neuroblastoma is the most common extra-cranial solid tumor in children and is metastatic at initial presentation in more than half of the cases. Although Neuroblastoma can be managed by surgery, chemotherapy, immunotherapy, and radiotherapy, its 5-year survival rate in children remains below 40%. Earlier studies have proposed the ketogenic diet as a possible adjuvant therapy for patients undergoing treatment for Neuroblastoma. In this study, we seek to review the possible roles of KD in the treatment of Neuroblastoma.
Collapse
Affiliation(s)
- Rangarirai Makuku
- Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Harare, Zimbabwe
| | - Zeinab Sinaei Far
- Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Neda Khalili
- Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alistar Moyo
- Universal Scientific Education and Research Network (USERN), Harare, Zimbabwe
| | - Sepideh Razi
- Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Nima Rezaei
- Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Stockholm, Sweden
| |
Collapse
|
26
|
Dialysis as a Novel Adjuvant Treatment for Malignant Cancers. Cancers (Basel) 2022; 14:cancers14205054. [PMID: 36291840 PMCID: PMC9600214 DOI: 10.3390/cancers14205054] [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: 09/09/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary There is a clear need for new cancer therapies as many cancers have a very short long-term survival rate. For most advanced cancers, therapy resistance limits the benefit of any single-agent chemotherapy, radiotherapy, or immunotherapy. Cancer cells show a greater dependence on glucose and glutamine as fuel than healthy cells do. In this article, we propose using 4- to 8-h dialysis treatments to change the blood composition, i.e., lowering glucose and glutamine levels, and elevating ketone levels—thereby disrupting major metabolic pathways important for cancer cell survival. The dialysis’ impact on cancer cells include not only metabolic effects, but also redox balance, immunological, and epigenetic effects. These pleiotropic effects could potentially enhance the effectiveness of traditional cancer treatments, such as radiotherapies, chemotherapies, and immunotherapies—resulting in improved outcomes and longer survival rates for cancer patients. Abstract Cancer metabolism is characterized by an increased utilization of fermentable fuels, such as glucose and glutamine, which support cancer cell survival by increasing resistance to both oxidative stress and the inherent immune system in humans. Dialysis has the power to shift the patient from a state dependent on glucose and glutamine to a ketogenic condition (KC) combined with low glutamine levels—thereby forcing ATP production through the Krebs cycle. By the force of dialysis, the cancer cells will be deprived of their preferred fermentable fuels, disrupting major metabolic pathways important for the ability of the cancer cells to survive. Dialysis has the potential to reduce glucose levels below physiological levels, concurrently increase blood ketone body levels and reduce glutamine levels, which may further reinforce the impact of the KC. Importantly, ketones also induce epigenetic changes imposed by histone deacetylates (HDAC) activity (Class I and Class IIa) known to play an important role in cancer metabolism. Thus, dialysis could be an impactful and safe adjuvant treatment, sensitizing cancer cells to traditional cancer treatments (TCTs), potentially making these significantly more efficient.
Collapse
|
27
|
Mitusova K, Peltek OO, Karpov TE, Muslimov AR, Zyuzin MV, Timin AS. Overcoming the blood-brain barrier for the therapy of malignant brain tumor: current status and prospects of drug delivery approaches. J Nanobiotechnology 2022; 20:412. [PMID: 36109754 PMCID: PMC9479308 DOI: 10.1186/s12951-022-01610-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/18/2022] [Indexed: 01/06/2023] Open
Abstract
Besides the broad development of nanotechnological approaches for cancer diagnosis and therapy, currently, there is no significant progress in the treatment of different types of brain tumors. Therapeutic molecules crossing the blood-brain barrier (BBB) and reaching an appropriate targeting ability remain the key challenges. Many invasive and non-invasive methods, and various types of nanocarriers and their hybrids have been widely explored for brain tumor treatment. However, unfortunately, no crucial clinical translations were observed to date. In particular, chemotherapy and surgery remain the main methods for the therapy of brain tumors. Exploring the mechanisms of the BBB penetration in detail and investigating advanced drug delivery platforms are the key factors that could bring us closer to understanding the development of effective therapy against brain tumors. In this review, we discuss the most relevant aspects of the BBB penetration mechanisms, observing both invasive and non-invasive methods of drug delivery. We also review the recent progress in the development of functional drug delivery platforms, from viruses to cell-based vehicles, for brain tumor therapy. The destructive potential of chemotherapeutic drugs delivered to the brain tumor is also considered. This review then summarizes the existing challenges and future prospects in the use of drug delivery platforms for the treatment of brain tumors.
Collapse
Affiliation(s)
- Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Oleksii O Peltek
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Albert R Muslimov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
- Sirius University of Science and Technology, Olympic Ave 1, Sirius, 354340, Russian Federation
| | - Mikhail V Zyuzin
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation.
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation.
| |
Collapse
|
28
|
Shen S, Iyengar NM. Insulin-Lowering Diets in Metastatic Cancer. Nutrients 2022; 14:nu14173542. [PMID: 36079800 PMCID: PMC9460605 DOI: 10.3390/nu14173542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Hyperinsulinemia is an independent risk factor for cancer mortality. Insulin-lowering dietary strategies such as calorie restriction (CR), low-carbohydrate or ketogenic diets (KD), and intermittent fasting (IF) are aimed at reducing systemic stores of nutrients utilized by cancer cells, attenuating insulin-related growth signaling, and improving obesity-related metabolic parameters. In this narrative review, we searched the published literature for studies that tested various insulin-lowering diets in metastatic cancer in preclinical and clinical settings. A total of 23 studies were identified. Of these, 14 were preclinical studies of dietary strategies that demonstrated improvements in insulin levels, inhibition of metastasis, and/or reduction in metastatic disease burden in animal models. The remaining nine clinical studies tested carbohydrate restriction, KD, or IF strategies which appear to be safe and feasible in patients with metastatic cancer. These approaches have also been shown to improve serum insulin and other metabolic parameters. Though promising, the anti-cancer efficacy of these interventions, such as impact on tumor response, disease-specific-, and overall survival, have not yet been conclusively demonstrated. Studies that are adequately powered to evaluate whether insulin-lowering diets improve cancer outcomes are warranted.
Collapse
Affiliation(s)
- Sherry Shen
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Neil M. Iyengar
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
29
|
Seyfried TN, Arismendi-Morillo G, Zuccoli G, Lee DC, Duraj T, Elsakka AM, Maroon JC, Mukherjee P, Ta L, Shelton L, D'Agostino D, Kiebish M, Chinopoulos C. Metabolic management of microenvironment acidity in glioblastoma. Front Oncol 2022; 12:968351. [PMID: 36059707 PMCID: PMC9428719 DOI: 10.3389/fonc.2022.968351] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma (GBM), similar to most cancers, is dependent on fermentation metabolism for the synthesis of biomass and energy (ATP) regardless of the cellular or genetic heterogeneity seen within the tumor. The transition from respiration to fermentation arises from the documented defects in the number, the structure, and the function of mitochondria and mitochondrial-associated membranes in GBM tissue. Glucose and glutamine are the major fermentable fuels that drive GBM growth. The major waste products of GBM cell fermentation (lactic acid, glutamic acid, and succinic acid) will acidify the microenvironment and are largely responsible for drug resistance, enhanced invasion, immunosuppression, and metastasis. Besides surgical debulking, therapies used for GBM management (radiation, chemotherapy, and steroids) enhance microenvironment acidification and, although often providing a time-limited disease control, will thus favor tumor recurrence and complications. The simultaneous restriction of glucose and glutamine, while elevating non-fermentable, anti-inflammatory ketone bodies, can help restore the pH balance of the microenvironment while, at the same time, providing a non-toxic therapeutic strategy for killing most of the neoplastic cells.
Collapse
Affiliation(s)
- Thomas N. Seyfried
- Biology Department, Boston College, Chestnut Hill, MA, United States
- *Correspondence: Thomas N. Seyfried,
| | - Gabriel Arismendi-Morillo
- Instituto de Investigaciones Biológicas, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Giulio Zuccoli
- The Program for the Study of Neurodevelopment in Rare Disorders (NDRD), University of Pittsburgh, Pittsburgh, PA, United States
| | - Derek C. Lee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Tomas Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, Madrid, Spain
| | - Ahmed M. Elsakka
- Neuro Metabolism, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Joseph C. Maroon
- Department of Neurosurgery, University of Pittsburgh, Medical Center, Pittsburgh, PA, United States
| | - Purna Mukherjee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Linh Ta
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | | | - Dominic D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | | | | |
Collapse
|
30
|
Clemente-Suárez VJ, Redondo-Flórez L, Rubio-Zarapuz A, Martínez-Guardado I, Navarro-Jiménez E, Tornero-Aguilera JF. Nutritional and Exercise Interventions in Cancer-Related Cachexia: An Extensive Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:4604. [PMID: 35457471 PMCID: PMC9025820 DOI: 10.3390/ijerph19084604] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 01/27/2023]
Abstract
One of the common traits found in cancer patients is malnutrition and cachexia, which affects between 25% to 60% of the patients, depending on the type of cancer, diagnosis, and treatment. Given the lack of current effective pharmacological solutions for low muscle mass and sarcopenia, holistic interventions are essential to patient care, as well as exercise and nutrition. Thus, the present narrative review aimed to analyze the nutritional, pharmacological, ergonutritional, and physical exercise strategies in cancer-related cachexia. The integration of multidisciplinary interventions could help to improve the final intervention in patients, improving their prognosis, quality of life, and life expectancy. To reach these aims, an extensive narrative review was conducted. The databases used were MedLine (PubMed), Cochrane (Wiley), Embase, PsychINFO, and CinAhl. Cancer-related cachexia is a complex multifactorial phenomenon in which systemic inflammation plays a key role in the development and maintenance of the symptomatology. Pharmacological interventions seem to produce a positive effect on inflammatory state and cachexia. Nutritional interventions are focused on a high-energy diet with high-density foods and the supplementation with antioxidants, while physical activity is focused on strength-based training. The implementation of multidisciplinary non-pharmacological interventions in cancer-related cachexia could be an important tool to improve traditional treatments and improve patients' quality of life.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (L.R.-F.); (A.R.-Z.); (J.F.T.-A.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Laura Redondo-Flórez
- Faculty of Sports Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (L.R.-F.); (A.R.-Z.); (J.F.T.-A.)
| | - Alejandro Rubio-Zarapuz
- Faculty of Sports Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (L.R.-F.); (A.R.-Z.); (J.F.T.-A.)
| | - Ismael Martínez-Guardado
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain;
| | | | | |
Collapse
|
31
|
Zhu H, Bi D, Zhang Y, Kong C, Du J, Wu X, Wei Q, Qin H. Ketogenic diet for human diseases: the underlying mechanisms and potential for clinical implementations. Signal Transduct Target Ther 2022; 7:11. [PMID: 35034957 PMCID: PMC8761750 DOI: 10.1038/s41392-021-00831-w] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/21/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
The ketogenic diet (KD) is a high-fat, adequate-protein, and very-low-carbohydrate diet regimen that mimics the metabolism of the fasting state to induce the production of ketone bodies. The KD has long been established as a remarkably successful dietary approach for the treatment of intractable epilepsy and has increasingly garnered research attention rapidly in the past decade, subject to emerging evidence of the promising therapeutic potential of the KD for various diseases, besides epilepsy, from obesity to malignancies. In this review, we summarize the experimental and/or clinical evidence of the efficacy and safety of the KD in different diseases, and discuss the possible mechanisms of action based on recent advances in understanding the influence of the KD at the cellular and molecular levels. We emphasize that the KD may function through multiple mechanisms, which remain to be further elucidated. The challenges and future directions for the clinical implementation of the KD in the treatment of a spectrum of diseases have been discussed. We suggest that, with encouraging evidence of therapeutic effects and increasing insights into the mechanisms of action, randomized controlled trials should be conducted to elucidate a foundation for the clinical use of the KD.
Collapse
Affiliation(s)
- Huiyuan Zhu
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Youhua Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Kong
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiahao Du
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Xiawei Wu
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinical College, Anhui Medical University, Hefei, China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Sevcikova A, Izoldova N, Stevurkova V, Kasperova B, Chovanec M, Ciernikova S, Mego M. The Impact of the Microbiome on Resistance to Cancer Treatment with Chemotherapeutic Agents and Immunotherapy. Int J Mol Sci 2022; 23:ijms23010488. [PMID: 35008915 PMCID: PMC8745082 DOI: 10.3390/ijms23010488] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 02/04/2023] Open
Abstract
Understanding the mechanisms of resistance to therapy in human cancer cells has become a multifaceted limiting factor to achieving optimal cures in cancer patients. Besides genetic and epigenetic alterations, enhanced DNA damage repair activity, deregulation of cell death, overexpression of transmembrane transporters, and complex interactions within the tumor microenvironment, other mechanisms of cancer treatment resistance have been recently proposed. In this review, we will summarize the preclinical and clinical studies highlighting the critical role of the microbiome in the efficacy of cancer treatment, concerning mainly chemotherapy and immunotherapy with immune checkpoint inhibitors. In addition to involvement in drug metabolism and immune surveillance, the production of microbiota-derived metabolites might represent the link between gut/intratumoral bacteria and response to anticancer therapies. Importantly, an emerging trend of using microbiota modulation by probiotics and fecal microbiota transplantation (FMT) to overcome cancer treatment resistance will be also discussed.
Collapse
Affiliation(s)
- Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská Cesta 9, 845 05 Bratislava, Slovakia; (A.S.); (N.I.); (V.S.)
| | - Nikola Izoldova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská Cesta 9, 845 05 Bratislava, Slovakia; (A.S.); (N.I.); (V.S.)
- Department of Genetics, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Viola Stevurkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská Cesta 9, 845 05 Bratislava, Slovakia; (A.S.); (N.I.); (V.S.)
| | - Barbora Kasperova
- Department of Oncohematology, Faculty of Medicine, Comenius University, Bratislava and National Cancer Institute, 833 10 Bratislava, Slovakia;
| | - Michal Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava and National Cancer Institute, 833 10 Bratislava, Slovakia; (M.C.); (M.M.)
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská Cesta 9, 845 05 Bratislava, Slovakia; (A.S.); (N.I.); (V.S.)
- Correspondence: ; Tel.: +421-2-3229-5198
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava and National Cancer Institute, 833 10 Bratislava, Slovakia; (M.C.); (M.M.)
| |
Collapse
|
33
|
Tasdogan A, Ubellacker JM, Morrison SJ. Redox Regulation in Cancer Cells during Metastasis. Cancer Discov 2021; 11:2682-2692. [PMID: 34649956 DOI: 10.1158/2159-8290.cd-21-0558] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/15/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
Metastasis is an inefficient process in which the vast majority of cancer cells are fated to die, partly because they experience oxidative stress. Metastasizing cancer cells migrate through diverse environments that differ dramatically from their tumor of origin, leading to redox imbalances. The rare metastasizing cells that survive undergo reversible metabolic changes that confer oxidative stress resistance. We review the changes in redox regulation that cancer cells undergo during metastasis. By better understanding these mechanisms, it may be possible to develop pro-oxidant therapies that block disease progression by exacerbating oxidative stress in cancer cells. SIGNIFICANCE: Oxidative stress often limits cancer cell survival during metastasis, raising the possibility of inhibiting cancer progression with pro-oxidant therapies. This is the opposite strategy of treating patients with antioxidants, an approach that worsened outcomes in large clinical trials.
Collapse
Affiliation(s)
- Alpaslan Tasdogan
- Children's Research Institute and Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jessalyn M Ubellacker
- Children's Research Institute and Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sean J Morrison
- Children's Research Institute and Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas. .,Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
34
|
Han P, Wang Y, Luo W, Lu Y, Zhou X, Yang Y, Zheng Q, Li D, Wu S, Li L, Zhang H, Zhao J, Zhang Z, Matskova L, Li P, Zhou X. Epigenetic inactivation of hydroxymethylglutaryl CoA synthase reduces ketogenesis and facilitates tumor cell motility in clear cell renal carcinoma. Pathol Res Pract 2021; 227:153622. [PMID: 34624592 DOI: 10.1016/j.prp.2021.153622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Previously, we have reported that the dysregulation of ketogenesis plays an important role in the carcinogenesis of clear cell renal cell carcinoma (ccRCC). Here, we demonstrate decreased expression of the HMGCS2 gene in ccRCC, a critical enzyme for the synthesis of the ketone body β-hydroxybutyrate (β-OHB). We found that the reduced transcription of the HMGCS2 gene in ccRCC cells was significantly correlated to a higher relative methylation rate in its promotor region. The higher methylation rate in the region of the transcription start site and 1st exon of the HMGCS2 gene was, in turn, correlated with a worse clinical outcome for patients. The transcription of HMGCS2 was possible to restore by treatment with 5-aza-2'-deoxycytidine and with the histone deacetylase inhibitor β-OHB. Therefore, the low levels of the HMGCS2 enzyme in ccRCC may be the consequence of hypermethylation of the HMGCS2 promotor. The ensuing reduction in the ketone body levels further suppresses the transcription of HMGCS2 via a feedback loop. Ectopic expression of HMGCS2 attenuates the migration and invasion of ccRCC but does not affect the proliferative capacity of ccRCC cells in vitro. In addition, we showed that ectopic expression of HMGCS2 boosts the intracellular levels of β-OHB and that exogenously applied β-OHB suppresses the motility and invasion of ccRCC. Our study reveals crosstalk between genes that regulate metabolism and their metabolites, thus providing a better understanding of the epigenetic mechanism involved in ccRCC carcinogenesis and suggesting opportunities for metabolic therapy of tumors. Initially, we suggest that the mRNA level of HMGCS2 could serve as a potentially valuable diagnostic (AUC = 0.918, p < 0.001) and prognostic biomarker.
Collapse
Affiliation(s)
- Peipei Han
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China; Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yifang Wang
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Wenqi Luo
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yunliang Lu
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Yanping Yang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Qian Zheng
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Danping Li
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Shu Wu
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Limei Li
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Haishan Zhang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Jun Zhao
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Zhe Zhang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Liudmila Matskova
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Ping Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Pathology, College & Hospital of Stomatology Guangxi Medical University, Nanning, China.
| | - Xiaoying Zhou
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China; Life Science Institute, Guangxi Medical University, Nanning, China.
| |
Collapse
|
35
|
Wang L, Chen P, Xiao W. β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients 2021; 13:nu13103420. [PMID: 34684426 PMCID: PMC8540704 DOI: 10.3390/nu13103420] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 12/18/2022] Open
Abstract
The ketone bodies, especially β-hydroxybutyrate (β-HB), derive from fatty acid oxidation and alternatively serve as a fuel source for peripheral tissues including the brain, heart, and skeletal muscle. β-HB is currently considered not solely an energy substrate for maintaining metabolic homeostasis but also acts as a signaling molecule of modulating lipolysis, oxidative stress, and neuroprotection. Besides, it serves as an epigenetic regulator in terms of histone methylation, acetylation, β-hydroxybutyrylation to delay various age-related diseases. In addition, studies support endogenous β-HB administration or exogenous supplementation as effective strategies to induce a metabolic state of nutritional ketosis. The purpose of this review article is to provide an overview of β-HB metabolism and its relationship and application in age-related diseases. Future studies are needed to reveal whether β-HB has the potential to serve as adjunctive nutritional therapy for aging.
Collapse
Affiliation(s)
| | - Peijie Chen
- Correspondence: (P.C.); (W.X.); Tel.: +86-021-65508039 (P.C.); +86-021-65507367 (W.X.)
| | - Weihua Xiao
- Correspondence: (P.C.); (W.X.); Tel.: +86-021-65508039 (P.C.); +86-021-65507367 (W.X.)
| |
Collapse
|
36
|
Wang L, Chen P, Xiao W. β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients 2021; 13:3420. [PMID: 34684426 PMCID: PMC8540704 DOI: 10.3390/nu13103420&set/a 930838900+926910489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
The ketone bodies, especially β-hydroxybutyrate (β-HB), derive from fatty acid oxidation and alternatively serve as a fuel source for peripheral tissues including the brain, heart, and skeletal muscle. β-HB is currently considered not solely an energy substrate for maintaining metabolic homeostasis but also acts as a signaling molecule of modulating lipolysis, oxidative stress, and neuroprotection. Besides, it serves as an epigenetic regulator in terms of histone methylation, acetylation, β-hydroxybutyrylation to delay various age-related diseases. In addition, studies support endogenous β-HB administration or exogenous supplementation as effective strategies to induce a metabolic state of nutritional ketosis. The purpose of this review article is to provide an overview of β-HB metabolism and its relationship and application in age-related diseases. Future studies are needed to reveal whether β-HB has the potential to serve as adjunctive nutritional therapy for aging.
Collapse
Affiliation(s)
| | - Peijie Chen
- Correspondence: (P.C.); (W.X.); Tel.: +86-021-65508039 (P.C.); +86-021-65507367 (W.X.)
| | - Weihua Xiao
- Correspondence: (P.C.); (W.X.); Tel.: +86-021-65508039 (P.C.); +86-021-65507367 (W.X.)
| |
Collapse
|
37
|
β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients 2021. [DOI: 10.3390/nu13103420
expr 933295879 + 814156476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
The ketone bodies, especially β-hydroxybutyrate (β-HB), derive from fatty acid oxidation and alternatively serve as a fuel source for peripheral tissues including the brain, heart, and skeletal muscle. β-HB is currently considered not solely an energy substrate for maintaining metabolic homeostasis but also acts as a signaling molecule of modulating lipolysis, oxidative stress, and neuroprotection. Besides, it serves as an epigenetic regulator in terms of histone methylation, acetylation, β-hydroxybutyrylation to delay various age-related diseases. In addition, studies support endogenous β-HB administration or exogenous supplementation as effective strategies to induce a metabolic state of nutritional ketosis. The purpose of this review article is to provide an overview of β-HB metabolism and its relationship and application in age-related diseases. Future studies are needed to reveal whether β-HB has the potential to serve as adjunctive nutritional therapy for aging.
Collapse
|
38
|
Cortez NE, Mackenzie GG. Ketogenic Diets in Pancreatic Cancer and Associated Cachexia: Cellular Mechanisms and Clinical Perspectives. Nutrients 2021; 13:nu13093202. [PMID: 34579079 PMCID: PMC8471358 DOI: 10.3390/nu13093202] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and extremely therapy-resistant cancer. It is estimated that up to 80% of PDAC patients present with cachexia, a multifactorial disorder characterized by the involuntary and ongoing wasting of skeletal muscle that affects therapeutic response and survival. During the last decade, there has been an increased interest in exploring dietary interventions to complement the treatment of PDAC and associated cachexia. Ketogenic diets (KDs) have gained attention for their anti-tumor potential. Characterized by a very low carbohydrate, moderate protein, and high fat composition, this diet mimics the metabolic changes that occur in fasting. Numerous studies report that a KD reduces tumor growth and can act as an adjuvant therapy in various cancers, including pancreatic cancer. However, research on the effect and mechanisms of action of KDs on PDAC-associated cachexia is limited. In this narrative review, we summarize the evidence of the impact of KDs in PDAC treatment and cachexia mitigation. Furthermore, we discuss key cellular mechanisms that explain KDs’ potential anti-tumor and anti-cachexia effects, focusing primarily on reprogramming of cell metabolism, epigenome, and the gut microbiome. Finally, we provide a perspective on future research needed to advance KDs into clinical use.
Collapse
|
39
|
Chapek MA, Martindale RG. Nutrition in Cancer Therapy: Overview for the Cancer Patient. JPEN J Parenter Enteral Nutr 2021; 45:33-40. [PMID: 34459006 DOI: 10.1002/jpen.2259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 11/10/2022]
Abstract
Despite significant advances in oncologic treatment, cancer-associated metabolic derangements remain largely poorly understood and often neglected in cancer care. Cancer cachexia and metabolic changes exhibited by neoplastic cells pose formidable barriers to improving outcomes and quality of life. Although cancer has traditionally been viewed as a proliferative disease caused by genetic mutations, newer perspectives suggest that it is primarily a metabolic disease. This paper discusses the etiology of cachexia and sarcopenia, and nutritional interventions that can address these wasting disorders. The role of inflammation in cancer and the methods for preventing and resolving inflammation with nutrition intervention are also explored. Several nutritional recommendations aimed at overcoming cachexia, resolving inflammation and improving cancer outcomes are provided based on current literature. This manuscript selected only a few areas in which to focus and is not all inclusive of the expansive literature available on the topic of cachexia. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Robert G Martindale
- Professor of Surgery, Division of GI and General Surgery, Oregon Health Sciences University, Portland, Oregon, United States
| |
Collapse
|
40
|
Ortega MA, Fraile-Martinez O, García-Montero C, Callejón-Peláez E, Sáez MA, Álvarez-Mon MA, García-Honduvilla N, Monserrat J, Álvarez-Mon M, Bujan J, Canals ML. A General Overview on the Hyperbaric Oxygen Therapy: Applications, Mechanisms and Translational Opportunities. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:864. [PMID: 34577787 PMCID: PMC8465921 DOI: 10.3390/medicina57090864] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022]
Abstract
Hyperbaric oxygen therapy (HBOT) consists of using of pure oxygen at increased pressure (in general, 2-3 atmospheres) leading to augmented oxygen levels in the blood (Hyperoxemia) and tissue (Hyperoxia). The increased pressure and oxygen bioavailability might be related to a plethora of applications, particularly in hypoxic regions, also exerting antimicrobial, immunomodulatory and angiogenic properties, among others. In this review, we will discuss in detail the physiological relevance of oxygen and the therapeutical basis of HBOT, collecting current indications and underlying mechanisms. Furthermore, potential areas of research will also be examined, including inflammatory and systemic maladies, COVID-19 and cancer. Finally, the adverse effects and contraindications associated with this therapy and future directions of research will be considered. Overall, we encourage further research in this field to extend the possible uses of this procedure. The inclusion of HBOT in future clinical research could be an additional support in the clinical management of multiple pathologies.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcala de Henares, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Enrique Callejón-Peláez
- Underwater and Hyperbaric Medicine Service, Central University Hospital of Defence—UAH Madrid, 28801 Alcala de Henares, Spain;
| | - Miguel A. Sáez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence—UAH Madrid, 28801 Alcala de Henares, Spain
| | - Miguel A. Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases—Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcala de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (N.G.-H.); (J.M.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - María Luisa Canals
- ISM, IMHA Research Chair, Former of IMHA (International Maritime Health Association), 43001 Tarragona, Spain;
| |
Collapse
|
41
|
Zhao B, Aggarwal A, Marshall JA, Barletta JA, Kijewski MF, Lorch JH, Nehs MA. Glycolytic inhibition with 3-bromopyruvate suppresses tumor growth and improves survival in a murine model of anaplastic thyroid cancer. Surgery 2021; 171:227-234. [PMID: 34334212 DOI: 10.1016/j.surg.2021.05.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Anaplastic thyroid cancer is a rare but devastating malignancy. Anaplastic thyroid cancer cells exhibit the Warburg effect by preferentially undergoing glycolysis even in aerobic conditions, leading to high glucose use. Here we assess if targeted inhibition of glycolysis can diminish anaplastic thyroid cancer growth and improve outcomes. METHODS Human anaplastic thyroid cancer cell line 8505C was grown in medium containing high (25 mmol/L) or low (3 mmol/L) glucose concentration and hexokinase II inhibitor 3-bromopyruvate (200 μM). Cellular proliferation, migration, and invasion were measured. An orthotopic xenograft model of anaplastic thyroid cancer was generated in nude mice using 8505C cells. Animals were provided standard chow or a ketogenic diet and treated with 3-bromopyruvate (1.8 mg/kg). Overall survival time was monitored. Necropsies were performed to harvest tumors for analysis. RESULTS Growth of 8505C in low-glucose medium with 3-bromopyruvate decreased cell proliferation by 89%, migration by 44%, and invasion by 73% (P < .001 for all) compared with high glucose. Animals concomitantly receiving a ketogenic diet and 3-bromopyruvate exhibited smaller tumor volumes (P = .03), slower tumor growth rates (P = .01), and improved overall survival (P = .006) compared with standard-diet control subjects. Monotherapy with a ketogenic diet or 3-bromopyruvate alone did not reduce tumor size or increase survival over the standard-diet control group. CONCLUSION Glycolytic inhibition with 3-bromopyruvate inhibits tumor growth and extends survival in a murine model of anaplastic thyroid cancer when combined with the ketogenic diet. Thus, targeted glycolytic inhibition of anaplastic thyroid cancer exhibits context-specific utility and may only be effective during ketosis induced by dietary restriction of glycolytic inputs.
Collapse
Affiliation(s)
- Bixiao Zhao
- Department of Surgery, Brigham and Women's Hospital, Boston MA. https://twitter.com/@BixiaoZhao
| | - Abha Aggarwal
- Department of Surgery, Brigham and Women's Hospital, Boston MA
| | | | | | - Marie F Kijewski
- Department of Radiology, Brigham and Women's Hospital, Boston MA
| | - Jochen H Lorch
- Head and Neck Center, Dana Farber Cancer Institute, Boston, MA. https://twitter.com/@DrLorch
| | - Matthew A Nehs
- Department of Surgery, Brigham and Women's Hospital, Boston MA.
| |
Collapse
|
42
|
Noble RJ, Walther V, Roumestand C, Hochberg ME, Hibner U, Lassus P. Paracrine Behaviors Arbitrate Parasite-Like Interactions Between Tumor Subclones. Front Ecol Evol 2021; 9. [PMID: 35096847 PMCID: PMC8794381 DOI: 10.3389/fevo.2021.675638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Explaining the emergence and maintenance of intratumor heterogeneity is an important question in cancer biology. Tumor cells can generate considerable subclonal diversity, which influences tumor growth rate, treatment resistance, and metastasis, yet we know remarkably little about how cells from different subclones interact. Here, we confronted two murine mammary cancer cell lines to determine both the nature and mechanisms of subclonal cellular interactions in vitro. Surprisingly, we found that, compared to monoculture, growth of the “winner” was enhanced by the presence of the “loser” cell line, whereas growth of the latter was reduced. Mathematical modeling and laboratory assays indicated that these interactions are mediated by the production of paracrine metabolites resulting in the winner subclone effectively “farming” the loser. Our findings add a new level of complexity to the mechanisms underlying subclonal growth dynamics.
Collapse
Affiliation(s)
- Robert J. Noble
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Department of Evolutionary Biology and Environmental Studies, University of Zurich, Zurich, Switzerland
- Correspondence: Patrice Lassus, Robert J. Noble
| | - Viola Walther
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Christian Roumestand
- Centre de Biochimie Structurale INSERM U1054, CNRS UMR 5048, Université de Montpellier, Montpellier, France
| | - Michael E. Hochberg
- Institute of Evolutionary Sciences, University of Montpellier, Montpellier, France
- Santa Fe Institute, Santa Fe, NM, United States
| | - Urszula Hibner
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Patrice Lassus
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
- Correspondence: Patrice Lassus, Robert J. Noble
| |
Collapse
|
43
|
Endicott M, Jones M, Hull J. Amino acid metabolism as a therapeutic target in cancer: a review. Amino Acids 2021; 53:1169-1179. [PMID: 34292410 PMCID: PMC8325646 DOI: 10.1007/s00726-021-03052-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/16/2021] [Indexed: 02/04/2023]
Abstract
Malignant cells often demonstrate a proliferative advantage when compared to non-malignant cells. However, the rapid growth and metabolism required for survival can also highlight vulnerabilities specific to these malignant cells. One such vulnerability exhibited by cancer is an increased demand for amino acids (AAs), which often results in a dependency on exogenous sources of AAs or requires upregulation of de novo synthesis. These metabolic alterations can be exploited by therapy, which aims to improve treatment outcome and decrease relapse and reoccurrence. One clinically utilised strategy targeting AA dependency is the use of asparaginase in the treatment of acute lymphoblastic leukaemia (ALL), which results in a depletion of exogenous asparagine and subsequent cancer cell death. Examples of other successful strategies include the exploitation of arginine deiminase and methioninase, nutrient restriction of methionine and the inhibition of glutaminase. In this review, we summarise these treatment strategies into three promising avenues: AA restriction, enzymatic depletion and inhibition of metabolism. This review provides an insight into the complexity of metabolism in cancer, whilst highlighting these three current research avenues that have support in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Molly Endicott
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Michael Jones
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Jonathon Hull
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
44
|
Schmidt DR, Patel R, Kirsch DG, Lewis CA, Vander Heiden MG, Locasale JW. Metabolomics in cancer research and emerging applications in clinical oncology. CA Cancer J Clin 2021; 71:333-358. [PMID: 33982817 PMCID: PMC8298088 DOI: 10.3322/caac.21670] [Citation(s) in RCA: 393] [Impact Index Per Article: 98.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer has myriad effects on metabolism that include both rewiring of intracellular metabolism to enable cancer cells to proliferate inappropriately and adapt to the tumor microenvironment, and changes in normal tissue metabolism. With the recognition that fluorodeoxyglucose-positron emission tomography imaging is an important tool for the management of many cancers, other metabolites in biological samples have been in the spotlight for cancer diagnosis, monitoring, and therapy. Metabolomics is the global analysis of small molecule metabolites that like other -omics technologies can provide critical information about the cancer state that are otherwise not apparent. Here, the authors review how cancer and cancer therapies interact with metabolism at the cellular and systemic levels. An overview of metabolomics is provided with a focus on currently available technologies and how they have been applied in the clinical and translational research setting. The authors also discuss how metabolomics could be further leveraged in the future to improve the management of patients with cancer.
Collapse
Affiliation(s)
- Daniel R. Schmidt
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708 USA
| | - Caroline A. Lewis
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Matthew G. Vander Heiden
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jason W. Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708 USA
| |
Collapse
|
45
|
Li J, Zhang H, Dai Z. Cancer Treatment With the Ketogenic Diet: A Systematic Review and Meta-analysis of Animal Studies. Front Nutr 2021; 8:594408. [PMID: 34179051 PMCID: PMC8219874 DOI: 10.3389/fnut.2021.594408] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/19/2021] [Indexed: 01/19/2023] Open
Abstract
Background: The ketogenic diet (KD) has been reported to play an important role in the development of cancer by an abundance of pre-clinical experiments; however, their conclusions have been controversial. We therefore aimed to perform a systematic review and meta-analysis of animal studies evaluating the effects of KD on cancer. Methods: Relevant studies were collected by searching PubMed, Embase, and Web of Science. Outcome measures comprised tumor weight, tumor volume, and survival time. Meta-analysis was performed using the random-effect model according to heterogeneity. Results: The search resulted in 1,254 references, of which 38 were included in the review and 17 included in the meta-analysis. Pooled results indicated that KD supplementation significantly prolonged survival time [standardized mean difference (SMD) = 1.76, 95% CI (0.58, 2.94), p = 0.003], and reduced tumor weight [SMD = -2.459, 95% CI (-4.188, -0.730), p = 0.027] and tumor volume [SMD = -0.759, 95% CI (-1.349, -0.168), p = 0.012]. Meta-regression and subgroup analysis results suggested that KD supplementation at a ratio of 4:1 was associated with remarkable prolongation of survival time in animals with limited tumor types. Conclusion: In summary, the pre-clinical evidence pointed toward an overall anti-tumor effect of the KD in animals studies currently available with limited tumor types.
Collapse
Affiliation(s)
- Jing Li
- Pharmaceutical Department, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyan Zhang
- Pharmaceutical Department, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhu Dai
- Pharmaceutical Department, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Sanchez-Pino MD, Gilmore LA, Ochoa AC, Brown JC. Obesity-Associated Myeloid Immunosuppressive Cells, Key Players in Cancer Risk and Response to Immunotherapy. Obesity (Silver Spring) 2021; 29:944-953. [PMID: 33616242 PMCID: PMC8154641 DOI: 10.1002/oby.23108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022]
Abstract
Obesity is a risk factor for developing several cancers. The dysfunctional metabolism and chronic activation of inflammatory pathways in obesity create a milieu that supports tumor initiation, progression, and metastasis. Obesity-associated metabolic, endocrine, and inflammatory mediators, besides interacting with cells leading to a malignant transformation, also modify the intrinsic metabolic and functional characteristics of immune myeloid cells. Here, the evidence supporting the hypothesis that obesity metabolically primes and promotes the expansion of myeloid cells with immunosuppressive and pro-oncogenic properties is discussed. In consequence, the accumulation of these cells, such as myeloid-derived suppressor cells and some subtypes of adipose-tissue macrophages, creates a microenvironment conducive to tumor development. In this review, the role of lipids, insulin, and leptin, which are dysregulated in obesity, is emphasized, as well as dietary nutrients in metabolic reprogramming of these myeloid cells. Moreover, emerging evidence indicating that obesity enhances immunotherapy response and hypothesized mechanisms are summarized. Priorities in deeper exploration involving the mechanisms of cross talk between metabolic disorders and myeloid cells related to cancer risk in patients with obesity are highlighted.
Collapse
Affiliation(s)
- Maria Dulfary Sanchez-Pino
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
- Department of Genetics, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
| | | | - Augusto C. Ochoa
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
- Department of Pediatrics, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
| | - Justin C. Brown
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
- Department of Genetics, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
- LSU Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| |
Collapse
|
47
|
Seyfried TN, Shivane AG, Kalamian M, Maroon JC, Mukherjee P, Zuccoli G. Ketogenic Metabolic Therapy, Without Chemo or Radiation, for the Long-Term Management of IDH1-Mutant Glioblastoma: An 80-Month Follow-Up Case Report. Front Nutr 2021; 8:682243. [PMID: 34136522 PMCID: PMC8200410 DOI: 10.3389/fnut.2021.682243] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Successful treatment of glioblastoma (GBM) remains futile despite decades of intense research. GBM is similar to most other malignant cancers in requiring glucose and glutamine for growth, regardless of histological or genetic heterogeneity. Ketogenic metabolic therapy (KMT) is a non-toxic nutritional intervention for cancer management. We report the case of a 32-year-old man who presented in 2014 with seizures and a right frontal lobe tumor on MRI. The tumor cells were immunoreactive with antibodies to the IDH1 (R132H) mutation, P53 (patchy), MIB-1 index (4–6%), and absent ATRX protein expression. DNA analysis showed no evidence of methylation of the MGMT gene promoter. The presence of prominent microvascular proliferation and areas of necrosis were consistent with an IDH-mutant glioblastoma (WHO Grade 4). Methods: The patient refused standard of care (SOC) and steroid medication after initial diagnosis, but was knowledgeable and self-motivated enough to consume a low-carbohydrate ketogenic diet consisting mostly of saturated fats, minimal vegetables, and a variety of meats. The patient used the glucose ketone index calculator to maintain his Glucose Ketone Index (GKI) near 2.0 without body weight loss. Results: The tumor continued to grow slowly without expected vasogenic edema until 2017, when the patient opted for surgical debulking. The enhancing area, centered in the inferior frontal gyrus, was surgically excised. The pathology specimen confirmed IDH1-mutant GBM. Following surgery, the patient continued with a self-administered ketogenic diet to maintain low GKI values, indicative of therapeutic ketosis. At the time of this report (May 2021), the patient remains alive with a good quality of life, except for occasional seizures. MRI continues to show slow interval progression of the tumor. Conclusion: This is the first report of confirmed IDH1-mutant GBM treated with KMT and surgical debulking without chemo- or radiotherapy. The long-term survival of this patient, now at 80 months, could be due in part to a therapeutic metabolic synergy between KMT and the IDH1 mutation that simultaneously target the glycolysis and glutaminolysis pathways that are essential for GBM growth. Further studies are needed to determine if this non-toxic therapeutic strategy could be effective in providing long-term management for other GBM patients with or without IDH mutations.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Aditya G Shivane
- Department of Cellular and Anatomical Pathology, University Hospital Plymouth National Health Service (NHS) Trust, Plymouth, United Kingdom
| | | | - Joseph C Maroon
- Department of Neurosurgery, Medical Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Purna Mukherjee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Giulio Zuccoli
- Department of Radiology, St. Christopher Hospital for Children, Drexel University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
48
|
The Chemistry of the Ketogenic Diet: Updates and Opportunities in Organic Synthesis. Int J Mol Sci 2021; 22:ijms22105230. [PMID: 34063366 PMCID: PMC8157195 DOI: 10.3390/ijms22105230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023] Open
Abstract
The high-fat, low-carbohydrate (ketogenic) diet has grown in popularity in the last decade as a weight loss tool. Research into the diet’s effects on the body have revealed a variety of other health benefits. The use of exogenous ketone supplements to confer the benefits of the diet without strict adherence to it represents an exciting new area of focus. Synthetic ketogenic compounds are of particular interest that has received very little emphasis and is an untapped area of focus for chemical synthesis. In this review, we summarize the chemical basis for ketogenicity and opportunities for further advancement of the field.
Collapse
|
49
|
Dowis K, Banga S. The Potential Health Benefits of the Ketogenic Diet: A Narrative Review. Nutrients 2021; 13:nu13051654. [PMID: 34068325 PMCID: PMC8153354 DOI: 10.3390/nu13051654] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/30/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023] Open
Abstract
Considering the lack of a comprehensive, multi-faceted overview of the ketogenic diet (KD) in relation to health issues, we compiled the evidence related to the use of the ketogenic diet in relation to its impact on the microbiome, the epigenome, diabetes, weight loss, cardiovascular health, and cancer. The KD diet could potentially increase genetic diversity of the microbiome and increase the ratio of Bacteroidetes to Firmicutes. The epigenome might be positively affected by the KD since it creates a signaling molecule known as β-hydroxybutyrate (BHB). KD has helped patients with diabetes reduce their HbA1c and reduce the need for insulin. There is evidence to suggest that a KD can help with weight loss, visceral adiposity, and appetite control. The evidence also suggests that eating a high-fat diet improves lipid profiles by lowering low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and lowering triglycerides (TG). Due to the Warburg effect, the KD is used as an adjuvant treatment to starve cancer cells, making them more vulnerable to chemotherapy and radiation. The potential positive impacts of a KD on each of these areas warrant further analysis, improved studies, and well-designed randomized controlled trials to further illuminate the therapeutic possibilities provided by this dietary intervention.
Collapse
|
50
|
Mierziak J, Burgberger M, Wojtasik W. 3-Hydroxybutyrate as a Metabolite and a Signal Molecule Regulating Processes of Living Organisms. Biomolecules 2021; 11:biom11030402. [PMID: 33803253 PMCID: PMC8000602 DOI: 10.3390/biom11030402] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
3-hydroxybutyrate (3-HB) as a very important metabolite occurs in animals, bacteria and plants. It is well known that in animals, 3-HB is formed as a product of the normal metabolism of fatty acid oxidation and can therefore be used as an energy source in the absence of sufficient blood glucose. In microorganisms, 3-HB mainly serves as a substrate for the synthesis of polyhydroxybutyrate, which is a reserve material. Recent studies show that in plants, 3-HB acts as a regulatory molecule that most likely influences the expression of genes involved in DNA methylation, thereby altering DNA methylation levels. Additionally, in animals, 3-HB is not only an intermediate metabolite, but also an important regulatory molecule that can influence gene expression, lipid metabolism, neuronal function, and overall metabolic rate. Some of these effects are the direct effects of 3-HB itself, while others are indirect effects, regulated by the metabolites into which 3-HB is converted. One of the most important regulatory functions of 3-HB is the inhibition of the activity of histone deacetylases and thus the epigenetic regulation of many genes. Due to the number of functions of this compound, it also shows promising therapeutic properties.
Collapse
|