1
|
Ligorio F, Vingiani A, Torelli T, Sposetti C, Drufuca L, Iannelli F, Zanenga L, Depretto C, Folli S, Scaperrotta G, Capri G, Bianchi GV, Ferraris C, Martelli G, Maugeri I, Provenzano L, Nichetti F, Agnelli L, Lobefaro R, Fucà G, Fotia G, Mariani L, Morelli D, Ladisa V, De Santis MC, Lozza L, Trecate G, Belfiore A, Brich S, Bertolotti A, Lorenzini D, Ficchì A, Martinetti A, Sottotetti E, Arata A, Corsetto P, Sorrentino L, Rediti M, Salvadori G, Minucci S, Foiani M, Apolone G, Pagani M, Pruneri G, de Braud F, Vernieri C. Early downmodulation of tumor glycolysis predicts response to fasting-mimicking diet in triple-negative breast cancer patients. Cell Metab 2024:S1550-4131(24)00450-9. [PMID: 39694040 DOI: 10.1016/j.cmet.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/04/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024]
Abstract
In preclinical experiments, cyclic fasting-mimicking diets (FMDs) showed broad anticancer effects in combination with chemotherapy. Among different tumor types, triple-negative breast cancer (TNBC) is exquisitely sensitive to FMD. However, the antitumor activity and efficacy of cyclic FMD in TNBC patients remain unclear. Here, we show that a severely calorie-restricted, triweekly, 5-day FMD regimen results in excellent pathologic complete response (pCR) rates (primary endpoint) and long-term clinical outcomes (secondary endpoints) when combined with preoperative chemotherapy in 30 patients with early-stage TNBC enrolled in the phase 2 trial BREAKFAST. Bulk and single-cell RNA sequencing analysis revealed that highly glycolytic cancer cells, myeloid cells, and pericytes from tumors achieving pCR undergo a significant, early downmodulation of pathways related to glycolysis and pyruvate metabolism. Our findings pave the wave for conducting larger clinical trials to investigate the efficacy of cyclic FMD in early-stage TNBC patients and to validate early changes of intratumor glycolysis as a predictor of clinical benefit from nutrient restriction. This study was registered at Clinicaltrials.gov (NCT04248998).
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Andrea Vingiani
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Tommaso Torelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Caterina Sposetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Lorenzo Drufuca
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Fabio Iannelli
- Haematopathogy Division, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Lucrezia Zanenga
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Catherine Depretto
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Secondo Folli
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Gianfranco Scaperrotta
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giuseppe Capri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giulia V Bianchi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Cristina Ferraris
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Gabriele Martelli
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Ilaria Maugeri
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Federico Nichetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Luca Agnelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giuseppe Fotia
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Luigi Mariani
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Daniele Morelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Vito Ladisa
- Hospital Pharmacy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Maria Carmen De Santis
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Laura Lozza
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Giovanna Trecate
- Department of Radiology and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Antonino Belfiore
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Silvia Brich
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Alessia Bertolotti
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Daniele Lorenzini
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Angela Ficchì
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Alessio Arata
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Paola Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Luca Sorrentino
- Surgical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Mattia Rediti
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giulia Salvadori
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Saverio Minucci
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Marco Foiani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Massimiliano Pagani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Giancarlo Pruneri
- Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy; Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; Oncology and Hematology-Oncology Department, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.
| |
Collapse
|
2
|
Islam MM, Sultana N, Liu C, Mao A, Katsube T, Wang B. Impact of dietary ingredients on radioprotection and radiosensitization: a comprehensive review. Ann Med 2024; 56:2396558. [PMID: 39320122 PMCID: PMC11425709 DOI: 10.1080/07853890.2024.2396558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
Radiation exposure poses significant health risks, particularly in radiotherapy and nuclear accidents. Certain dietary ingredients offer potential radioprotection and radiosensitization. In this review, we explore the impact of dietary ingredients, including vitamins, minerals, antioxidants, and other bioactive compounds, on radiation sensitivity and their potential for radioprotection. Radiosensitizers reoxygenate hypoxic tumor cells, increase the radiolysis of water molecules, and regulate various molecular mechanisms to induce cytotoxicity and inhibit DNA repair in irradiated tumor cells. Several dietary ingredients, such as vitamins C, E, selenium, and phytochemicals, show promise in protecting against radiation by reducing radiation-induced oxidative stress, inflammation, and DNA damage. Radioprotectors, such as ascorbic acid, curcumin, resveratrol, and genistein, activate and modulate various signaling pathways, including Keap1-Nrf2, NF-κB, PI3K/Akt/mammalian target of rapamycin (mTOR), STAT3, and mitogen-activated protein kinase (MAPK), in response to radiation-induced oxidative stress, regulating inflammatory cytokine expression, and promoting DNA damage repair and cell survival. Conversely, natural dietary radiosensitizers impede these pathways by enhancing DNA damage and inducing apoptosis in irradiated tumor cells. Understanding the molecular basis of these effects may aid in the development of effective strategies for radioprotection and radiosensitization in cancer treatment. Dietary interventions have the potential to enhance the efficacy of radiation therapy and minimize the side effects associated with radiation exposure.
Collapse
Affiliation(s)
- Md Monirul Islam
- Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka, Bangladesh
| | - Nahida Sultana
- Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka, Bangladesh
| | - Chang Liu
- Department of Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Aihong Mao
- Center of Medical Molecular Biology Research, Gansu Provincial Cancer Hospital, Gansu Provincial Academic Institute for Medical Research, Lanzhou, PR China
| | - Takanori Katsube
- Institute for Radiological Science, Quantum Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Bing Wang
- Institute for Radiological Science, Quantum Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| |
Collapse
|
3
|
Tagliafico L, Da Costa RT, Boccia L, Kavehmoghaddam S, Ramirez B, Tokarska‐Schlattner M, Scoma ER, Hambardikar V, Bonfiglio T, Caffa I, Monacelli F, Schlattner U, Betley JN, Nencioni A, Solesio ME. Short-term starvation activates AMPK and restores mitochondrial inorganic polyphosphate, but fails to reverse associated neuronal senescence. Aging Cell 2024; 23:e14289. [PMID: 39102875 PMCID: PMC11561667 DOI: 10.1111/acel.14289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024] Open
Abstract
Neuronal senescence is a major risk factor for the development of many neurodegenerative disorders. The mechanisms that drive neurons to senescence remain largely elusive; however, dysregulated mitochondrial physiology seems to play a pivotal role in this process. Consequently, strategies aimed to preserve mitochondrial function may hold promise in mitigating neuronal senescence. For example, dietary restriction has shown to reduce senescence, via a mechanism that still remains far from being totally understood, but that could be at least partially mediated by mitochondria. Here, we address the role of mitochondrial inorganic polyphosphate (polyP) in the intersection between neuronal senescence and dietary restriction. PolyP is highly present in mammalian mitochondria; and its regulatory role in mammalian bioenergetics has already been described by us and others. Our data demonstrate that depletion of mitochondrial polyP exacerbates neuronal senescence, independently of whether dietary restriction is present. However, dietary restriction in polyP-depleted cells activates AMPK, and it restores some components of mitochondrial physiology, even if this is not sufficient to revert increased senescence. The effects of dietary restriction on polyP levels and AMPK activation are conserved in differentiated SH-SY5Y cells and brain tissue of male mice. Our results identify polyP as an important component in mitochondrial physiology at the intersection of dietary restriction and senescence, and they highlight the importance of the organelle in this intersection.
Collapse
Affiliation(s)
- Luca Tagliafico
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
- Department of Internal Medicine and Medical Specialties (DIMI)University of GenoaGenoaItaly
| | - Renata T. Da Costa
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
| | - Lavinia Boccia
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sheida Kavehmoghaddam
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
| | - Bryan Ramirez
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
| | | | - Ernest R. Scoma
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
| | - Vedangi Hambardikar
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
| | - Tommaso Bonfiglio
- Department of Internal Medicine and Medical Specialties (DIMI)University of GenoaGenoaItaly
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties (DIMI)University of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI)University of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied BioenergeticsGrenoble Alpes UniversitySaint‐Martin‐d'HèresFrance
| | - J. Nicholas Betley
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI)University of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Maria E. Solesio
- Department of Biology and Center for Computational and Integrative BiologyRutgers UniversityCamdenNew JerseyUSA
| |
Collapse
|
4
|
Fulton TL, Johnstone JN, Tan JJ, Balagopal K, Dedman A, Chan AY, Johnson TK, Mirth CK, Piper MDW. Transiently restricting individual amino acids protects Drosophila melanogaster against multiple stressors. Open Biol 2024; 14:240093. [PMID: 39106944 PMCID: PMC11303031 DOI: 10.1098/rsob.240093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 08/09/2024] Open
Abstract
Nutrition and resilience are linked, though it is not yet clear how diet confers stress resistance or the breadth of stressors that it can protect against. We have previously shown that transiently restricting an essential amino acid can protect Drosophila melanogaster against nicotine poisoning. Here, we sought to characterize the nature of this dietary-mediated protection and determine whether it was sex, amino acid and/or nicotine specific. When we compared between sexes, we found that isoleucine deprivation increases female, but not male, nicotine resistance. Surprisingly, we found that this protection afforded to females was not replicated by dietary protein restriction and was instead specific to individual amino acid restriction. To understand whether these beneficial effects of diet were specific to nicotine or were generalizable across stressors, we pre-treated flies with amino acid restriction diets and exposed them to other types of stress. We found that some of the diets that protected against nicotine also protected against oxidative and starvation stress, and improved survival following cold shock. Interestingly, we found that a diet lacking isoleucine was the only diet to protect against all these stressors. These data point to isoleucine as a critical determinant of robustness in the face of environmental challenges.
Collapse
Affiliation(s)
- Tahlia L. Fulton
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Joshua N. Johnstone
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Jing J. Tan
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Krithika Balagopal
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Amy Dedman
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Andrea Y. Chan
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria3800, Australia
| | - Travis K. Johnson
- School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria3086, Australia
| | - Christen K. Mirth
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| | - Matthew D. W. Piper
- School of Biological Sciences, Monash University, Clayton, Victoria3800, Australia
| |
Collapse
|
5
|
Raucci F, Vernieri C, Di Tano M, Ligorio F, Blaževitš O, Lazzeri S, Shmahala A, Fragale G, Salvadori G, Varano G, Casola S, Buono R, Visco E, de Braud F, Longo VD. Cyclic Fasting-Mimicking Diet Plus Bortezomib and Rituximab Is an Effective Treatment for Chronic Lymphocytic Leukemia. Cancer Res 2024; 84:1133-1148. [PMID: 38241703 PMCID: PMC10982641 DOI: 10.1158/0008-5472.can-23-0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 10/12/2023] [Accepted: 01/18/2024] [Indexed: 01/21/2024]
Abstract
Cyclic fasting-mimicking diet (FMD) is an experimental nutritional intervention with potent antitumor activity in preclinical models of solid malignancies. FMD cycles are also safe and active metabolically and immunologically in cancer patients. Here, we reported on the outcome of FMD cycles in two patients with chronic lymphocytic leukemia (CLL) and investigated the effects of fasting and FMD cycles in preclinical CLL models. Fasting-mimicking conditions in murine CLL models had mild cytotoxic effects, which resulted in apoptosis activation mediated in part by lowered insulin and IGF1 concentrations. In CLL cells, fasting conditions promoted an increase in proteasome activity that served as a starvation escape pathway. Pharmacologic inhibition of this escape mechanism with the proteasome inhibitor bortezomib resulted in a strong enhancement of the proapoptotic effects of starvation conditions in vitro. In mouse CLL models, combining cyclic fasting/FMD with bortezomib and rituximab, an anti-CD20 antibody, delayed CLL progression and resulted in significant prolongation of mouse survival. Overall, the effect of proteasome inhibition in combination with FMD cycles in promoting CLL death supports the targeting of starvation escape pathways as an effective treatment strategy that should be tested in clinical trials. SIGNIFICANCE Chronic lymphocytic leukemia cells resist fasting-mimicking diet by inducing proteasome activation to escape starvation, which can be targeted using proteasome inhibition by bortezomib treatment to impede leukemia progression and prolong survival.
Collapse
Affiliation(s)
- Franca Raucci
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Claudio Vernieri
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maira Di Tano
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Weill Cornell Medical College, Department of Medicine, Cornell University, New York, New York
| | - Francesca Ligorio
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Olga Blaževitš
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Samuel Lazzeri
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Giuseppe Fragale
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giulia Salvadori
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Gabriele Varano
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Stefano Casola
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Roberta Buono
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
- Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, California
| | - Euplio Visco
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valter D. Longo
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, California
| |
Collapse
|
6
|
Nemati M, Shayanfar M, Almasi F, Mohammad-Shirazi M, Sharifi G, Aminianfar A, Esmaillzadeh A. Dietary patterns in relation to glioma: a case-control study. Cancer Metab 2024; 12:8. [PMID: 38500219 PMCID: PMC10946126 DOI: 10.1186/s40170-024-00336-4] [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: 11/18/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Although the association of individual foods and nutrients with glioma have been investigated, studies on the association of major dietary patterns and glioma are scarce. The aim of this study was to examine the association between major dietary patterns and risk of glioma in a group of Iranian adults. In this hospital-based case-control design, we recruited 128 newly diagnosed glioma cases and 256 controls in Tehran from 2009 to 2011. A Willett-format-validated 126-item semi-quantitative Food Frequency Questionnaire (FFQ) was used to assess participants' dietary intake. Factor analysis was used to identify major dietary patterns. We identified 3 major dietary patterns using factor analysis: high protein, vegetarian and western dietary pattern. After several adjustments for potential confounders, adherence to the high protein dietary pattern was inversely associated with risk of glioma (OR: 0.47; 95% CI: 0.23, 0.95). Consumption of vegetarian dietary pattern was also associated with a reduced risk of glioma (OR: 0.16; 95% CI: 0.07, 0.34). Greater adherence to the western dietary pattern was associated with a greater chance of glioma (OR: 3.30; 95% CI: 1.52, 7.17). We found that high protein, vegetarian and western dietary pattern were significantly associated with glioma risk. Further prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Mohammad Nemati
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shayanfar
- Department of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Almasi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Science, Kashan University of Medical Sciences, No. 226, Ravand Blv, Kashan, 1416753955, Iran
| | - Minoo Mohammad-Shirazi
- Department of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Giuve Sharifi
- Department of Neurosurgery, Loghman Hakim Hospital, Tehran, Iran
| | - Azadeh Aminianfar
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Science, Kashan University of Medical Sciences, No. 226, Ravand Blv, Kashan, 1416753955, Iran.
| | - Ahmad Esmaillzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
7
|
Margetis AT. Caloric restriction for the management of malignant tumors - from animal studies towards clinical translation. INT J VITAM NUTR RES 2024; 94:1-9. [PMID: 36755497 DOI: 10.1024/0300-9831/a000779] [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] [Indexed: 02/10/2023]
Abstract
In the last few years, numerous studies have demonstrated that dietary modifications in the form of calory restriction exert beneficial effects in several clinical entities, including aging-related pathologies, autoimmune diseases and cancer. Both as preventive but also as therapeutic modalities, these dietary regimens can impact systemic metabolism, immune and hormonal responses, redox balance and gut microbiota, among others. In the field of oncology, the vast majority of experimental work has explored the role of restricted diets in the prevention of malignant tumors, mostly in carcinogenesis-induced models, with at least encouraging results; on the contrary, less research has been performed in the management of full-blown cancer with ketogenic diet or caloric restriction protocols. Herein, we are aiming to review the relevant preclinical and clinical studies to date that investigate the role of caloric restriction in the treatment of established cancer.
Collapse
Affiliation(s)
- Aggelos T Margetis
- Internal Medicine-Oncology Residency Program, 2nd Department of Internal Medicine, Naval and Veterans Hospital, Athens, Greece
| |
Collapse
|
8
|
Mishra A, Giuliani G, Longo VD. Nutrition and dietary restrictions in cancer prevention. Biochim Biophys Acta Rev Cancer 2024; 1879:189063. [PMID: 38147966 DOI: 10.1016/j.bbcan.2023.189063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/15/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
The composition and pattern of dietary intake have emerged as key factors influencing aging, regeneration, and consequently, healthspan and lifespan. Cancer is one of the major diseases more tightly linked with aging, and age-related mortality. Although the role of nutrition in cancer incidence is generally well established, we are far from a consensus on how diet influences tumour development in different tissues. In this review, we will discuss how diet and dietary restrictions affect cancer risk and the molecular mechanisms potentially responsible for their effects. We will cover calorie restriction, intermittent fasting, prolonged fasting, fasting-mimicking diet, time-restricted eating, ketogenic diet, high protein diet, Mediterranean diet, and the vegan and vegetarian diets.
Collapse
Affiliation(s)
- Amrendra Mishra
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Giacomo Giuliani
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Valter D Longo
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; IFOM, FIRC Institute of Molecular Oncology, Via Adamello, 16, 20139 Milano, Italy.
| |
Collapse
|
9
|
Buono R, Tucci J, Cutri R, Guidi N, Mangul S, Raucci F, Pellegrini M, Mittelman SD, Longo VD. Fasting-Mimicking Diet Inhibits Autophagy and Synergizes with Chemotherapy to Promote T-Cell-Dependent Leukemia-Free Survival. Cancers (Basel) 2023; 15:5870. [PMID: 38136414 PMCID: PMC10741737 DOI: 10.3390/cancers15245870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Fasting mimicking diets (FMDs) are effective in the treatment of many solid tumors in mouse models, but their effect on hematologic malignancies is poorly understood, particularly in combination with standard therapies. Here we show that cycles of a 3-day FMD given to high-fat-diet-fed mice once a week increased the efficacy of vincristine to improve survival from BCR-ABL B acute lymphoblastic leukemia (ALL). In mice fed a standard diet, FMD cycles in combination with vincristine promoted cancer-free survival. RNA seq and protein assays revealed a vincristine-dependent decrease in the expression of multiple autophagy markers, which was exacerbated by the fasting/FMD conditions. The autophagy inhibitor chloroquine could substitute for fasting/FMD to promote cancer-free survival in combination with vincristine. In vitro, targeted inhibition of autophagy genes ULK1 and ATG9a strongly potentiated vincristine's toxicity. Moreover, anti-CD8 antibodies reversed the effects of vincristine plus fasting/FMD in promoting leukemia-free survival in mice, indicating a central role of the immune system in this response. Thus, the inhibition of autophagy and enhancement of immune responses appear to be mediators of the fasting/FMD-dependent cancer-free survival in ALL mice.
Collapse
Affiliation(s)
- Roberta Buono
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Jonathan Tucci
- Center for Endocrinology, Diabetes & Metabolism, Children’s Hospital Los Angeles, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Raffaello Cutri
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
| | - Novella Guidi
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
| | - Serghei Mangul
- Department of Computer Science, University of California Los Angeles, 580 Portola Plaza, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, Boyer Hall, 611 Charles Young Drive, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Franca Raucci
- IFOM AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Matteo Pellegrini
- Institute for Quantitative and Computational Biosciences, Boyer Hall, 611 Charles Young Drive, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, 801 Hilgard Avenue, Los Angeles, CA 90095, USA
| | - Steven D. Mittelman
- Center for Endocrinology, Diabetes & Metabolism, Children’s Hospital Los Angeles, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
- Division of Pediatric Endocrinology, UCLA Mattel Children’s Hospital, 10833 Le Conte Avenue, MDCC 22-315, Los Angeles, CA 90095, USA
| | - Valter D. Longo
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
- IFOM AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
10
|
Becker S, Momoh J, Biancacci I, Möckel D, Wang Q, May JN, Su H, Candels LS, Berres ML, Kiessling F, Hatting M, Lammers T, Trautwein C. Intermittent Fasting Primes the Tumor Microenvironment and Improves Nanomedicine Delivery in Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208042. [PMID: 37376850 DOI: 10.1002/smll.202208042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/18/2023] [Indexed: 06/29/2023]
Abstract
Fasting has many health benefits, including reduced chemotherapy toxicity and improved efficacy. It is unclear how fasting affects the tumor microenvironment (TME) and tumor-targeted drug delivery. Here the effects of intermittent (IF) and short-term (STF) fasting are investigated on tumor growth, TME composition, and liposome delivery in allogeneic hepatocellular carcinoma (HCC) mouse models. To this end, mice are inoculated either subcutaneously or intrahepatically with Hep-55.1C cells and subjected to IF for 24 d or to STF for 1 d. IF but not STF significantly slows down tumor growth. IF increases tumor vascularization and decreases collagen density, resulting in improved liposome delivery. In vitro, fasting furthermore promotes the tumor cell uptake of liposomes. These results demonstrate that IF shapes the TME in HCC towards enhanced drug delivery. Finally, when combining IF with liposomal doxorubicin treatment, the antitumor efficacy of nanochemotherapy is found to be increased, while systemic side effects are reduced. Altogether, these findings exemplify that the beneficial effects of fasting on anticancer therapy outcomes go beyond modulating metabolism at the molecular level.
Collapse
Affiliation(s)
- Svea Becker
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Jeffrey Momoh
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Ilaria Biancacci
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Diana Möckel
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Qingbi Wang
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Jan-Niklas May
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Huan Su
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Lena Susanna Candels
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Marie-Luise Berres
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Maximilian Hatting
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Christian Trautwein
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| |
Collapse
|
11
|
Cherkasova V, Ilnytskyy Y, Kovalchuk O, Kovalchuk I. Transcriptome Analysis of Cisplatin, Cannabidiol, and Intermittent Serum Starvation Alone and in Various Combinations on Colorectal Cancer Cells. Int J Mol Sci 2023; 24:14743. [PMID: 37834191 PMCID: PMC10572413 DOI: 10.3390/ijms241914743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Platinum-derived chemotherapy medications are often combined with other conventional therapies for treating different tumors, including colorectal cancer. However, the development of drug resistance and multiple adverse effects remain common in clinical settings. Thus, there is a necessity to find novel treatments and drug combinations that could effectively target colorectal cancer cells and lower the probability of disease relapse. To find potential synergistic interaction, we designed multiple different combinations between cisplatin, cannabidiol, and intermittent serum starvation on colorectal cancer cell lines. Based on the cell viability assay, we found that combinations between cannabidiol and intermittent serum starvation, cisplatin and intermittent serum starvation, as well as cisplatin, cannabidiol, and intermittent serum starvation can work in a synergistic fashion on different colorectal cancer cell lines. Furthermore, we analyzed differentially expressed genes and affected pathways in colorectal cancer cell lines to understand further the potential molecular mechanisms behind the treatments and their interactions. We found that synergistic interaction between cannabidiol and intermittent serum starvation can be related to changes in the transcription of genes responsible for cell metabolism and cancer's stress pathways. Moreover, when we added cisplatin to the treatments, there was a strong enrichment of genes taking part in G2/M cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
| | | | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (V.C.); (Y.I.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (V.C.); (Y.I.)
| |
Collapse
|
12
|
Jiménez-Alonso JJ, López-Lázaro M. Dietary Manipulation of Amino Acids for Cancer Therapy. Nutrients 2023; 15:2879. [PMID: 37447206 DOI: 10.3390/nu15132879] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer cells cannot proliferate and survive unless they obtain sufficient levels of the 20 proteinogenic amino acids (AAs). Unlike normal cells, cancer cells have genetic and metabolic alterations that may limit their capacity to obtain adequate levels of the 20 AAs in challenging metabolic environments. However, since normal diets provide all AAs at relatively constant levels and ratios, these potentially lethal genetic and metabolic defects are eventually harmless to cancer cells. If we temporarily replace the normal diet of cancer patients with artificial diets in which the levels of specific AAs are manipulated, cancer cells may be unable to proliferate and survive. This article reviews in vivo studies that have evaluated the antitumor activity of diets restricted in or supplemented with the 20 proteinogenic AAs, individually and in combination. It also reviews our recent studies that show that manipulating the levels of several AAs simultaneously can lead to marked survival improvements in mice with metastatic cancers.
Collapse
Affiliation(s)
| | - Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Sevilla, Spain
| |
Collapse
|
13
|
Ferro Y, Maurotti S, Tarsitano MG, Lodari O, Pujia R, Mazza E, Lascala L, Russo R, Pujia A, Montalcini T. Therapeutic Fasting in Reducing Chemotherapy Side Effects in Cancer Patients: A Systematic Review and Meta-Analysis. Nutrients 2023; 15:2666. [PMID: 37375570 PMCID: PMC10303481 DOI: 10.3390/nu15122666] [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: 04/26/2023] [Revised: 05/12/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The aim of this study was to assess the available evidence regarding the effect of a variety of fasting-like regimens on preventing chemotherapy-related side effects. PubMed, Scopus and Embase were used to select the studies for this review, which concluded on 24 November 2022. All types of clinical trials and case series reporting chemotherapy toxicity associated with fasting regimens and any comparison were considered. A total of 283 records were identified, of which 274 were excluded, leaving only nine studies that met the inclusion criteria. Five of these trials were randomized. Overall, moderate to high-quality evidence showed that several fasting regimens did not provide benefits compared to a conventional diet or other comparators in reducing the risk of adverse events. The overall pooled estimate for a variety of fasting regime when compared to non-fasting, indicated no significant difference in the side effects (RR = 1.10; 95% CI: 0.77-1.59; I2 = 10%, p = 0.60), including neutropenia alone (RR = 1.33; 95% CI: 0.90-1.97; I2 = 0%, p = 0.15). A sensitivity analysis confirmed these results. Based on our systematic review and meta-analysis, there is currently no evidence supporting the superiority of therapeutic fasting over non-fasting in preventing chemotherapy toxicity. The development of cancer treatment that do not entail toxicities remains imperative.
Collapse
Affiliation(s)
- Yvelise Ferro
- Department of Medical and Surgical Science, University Magna Grecia, 88100 Catanzaro, Italy; (Y.F.); (M.G.T.); (R.P.); (R.R.); (A.P.)
| | - Samantha Maurotti
- Department of Clinical and Experimental Medicine, University Magna Grecia, 88100 Catanzaro, Italy; (S.M.); (O.L.); (L.L.); (T.M.)
| | - Maria Grazia Tarsitano
- Department of Medical and Surgical Science, University Magna Grecia, 88100 Catanzaro, Italy; (Y.F.); (M.G.T.); (R.P.); (R.R.); (A.P.)
| | - Oscar Lodari
- Department of Clinical and Experimental Medicine, University Magna Grecia, 88100 Catanzaro, Italy; (S.M.); (O.L.); (L.L.); (T.M.)
| | - Roberta Pujia
- Department of Medical and Surgical Science, University Magna Grecia, 88100 Catanzaro, Italy; (Y.F.); (M.G.T.); (R.P.); (R.R.); (A.P.)
| | - Elisa Mazza
- Department of Medical and Surgical Science, University Magna Grecia, 88100 Catanzaro, Italy; (Y.F.); (M.G.T.); (R.P.); (R.R.); (A.P.)
| | - Lidia Lascala
- Department of Clinical and Experimental Medicine, University Magna Grecia, 88100 Catanzaro, Italy; (S.M.); (O.L.); (L.L.); (T.M.)
| | - Raffaella Russo
- Department of Medical and Surgical Science, University Magna Grecia, 88100 Catanzaro, Italy; (Y.F.); (M.G.T.); (R.P.); (R.R.); (A.P.)
| | - Arturo Pujia
- Department of Medical and Surgical Science, University Magna Grecia, 88100 Catanzaro, Italy; (Y.F.); (M.G.T.); (R.P.); (R.R.); (A.P.)
- Research Center for the Prevention and Treatment of Metabolic Diseases, University Magna Grecia, 88100 Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, University Magna Grecia, 88100 Catanzaro, Italy; (S.M.); (O.L.); (L.L.); (T.M.)
- Research Center for the Prevention and Treatment of Metabolic Diseases, University Magna Grecia, 88100 Catanzaro, Italy
| |
Collapse
|
14
|
Wünsch AC, Ries E, Heinzelmann S, Frabschka A, Wagner PC, Rauch T, Koderer C, El-Mesery M, Volland JM, Kübler AC, Hartmann S, Seher A. Metabolic Silencing via Methionine-Based Amino Acid Restriction in Head and Neck Cancer. Curr Issues Mol Biol 2023; 45:4557-4573. [PMID: 37367038 DOI: 10.3390/cimb45060289] [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: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
In recent years, various forms of caloric restriction (CR) and amino acid or protein restriction (AAR or PR) have shown not only success in preventing age-associated diseases, such as type II diabetes and cardiovascular diseases, but also potential for cancer therapy. These strategies not only reprogram metabolism to low-energy metabolism (LEM), which is disadvantageous for neoplastic cells, but also significantly inhibit proliferation. Head and neck squamous cell carcinoma (HNSCC) is one of the most common tumour types, with over 600,000 new cases diagnosed annually worldwide. With a 5-year survival rate of approximately 55%, the poor prognosis has not improved despite extensive research and new adjuvant therapies. Therefore, for the first time, we analysed the potential of methionine restriction (MetR) in selected HNSCC cell lines. We investigated the influence of MetR on cell proliferation and vitality, the compensation for MetR by homocysteine, the gene regulation of different amino acid transporters, and the influence of cisplatin on cell proliferation in different HNSCC cell lines.
Collapse
Affiliation(s)
- Anna Chiara Wünsch
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Elena Ries
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Sina Heinzelmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Andrea Frabschka
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Peter Christoph Wagner
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Theresa Rauch
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Corinna Koderer
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Julian Manuel Volland
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Alexander Christian Kübler
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Stefan Hartmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| | - Axel Seher
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
| |
Collapse
|
15
|
Farshbaf M, Mojarad-Jabali S, Hemmati S, Khosroushahi AY, Motasadizadeh H, Zarebkohan A, Valizadeh H. Enhanced BBB and BBTB penetration and improved anti-glioma behavior of Bortezomib through dual-targeting nanostructured lipid carriers. J Control Release 2022; 345:371-384. [PMID: 35301054 DOI: 10.1016/j.jconrel.2022.03.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/12/2022] [Accepted: 03/10/2022] [Indexed: 12/19/2022]
Abstract
The effective treatment of glioma through conventional chemotherapy is proved to be a great challenge in clinics. The main reason is due to the existence of two physiological and pathological barriers respectively including the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) that prevent most of the chemotherapeutics from efficient delivery to the brain tumors. To address this challenge, an ideal drug delivery system would efficiently traverse the BBB and BBTB and deliver the therapeutics into the glioma cells with high selectivity. Herein, a targeted delivery system was developed based on nanostructured lipid carriers (NLCs) modified with two proteolytically stable D-peptides, D8 and RI-VAP (Dual NLCs). D8 possesses high affinity towards nicotine acetylcholine receptors (nAChRs), overexpressed on brain capillary endothelial cells (BCECs), and can penetrate through BBB with high efficiency. RI-VAP is a specific ligand of cell surface GRP78 (csGRP78), a specific angiogenesis and cancer cell-surface marker, capable of circumventing the BBTB with superior glioma-homing property. Dual NLCs could internalize into BCECs, tumor neovascular endothelial cells, and glioma cells with high specificity and could penetrate through in vitro BBB and BBTB models with excellent efficiency compared to non-targeted or mono-targeted NLCs. In vivo whole-animal imaging and ex vivo imaging further confirmed the superior targeting capability of Dual NLCs towards intracranial glioma. When loaded with Bortezomib (BTZ), Dual NLCs attained the highest therapeutic efficiency by means of in vitro cytotoxicity and apoptosis and prolonged survival rate and anti-glioma behavior in intracranial glioma bearing mice. Collectively, the designed targeting platform in this study could overcome multiple barriers and effectively deliver BTZ to glioma cells, which represent its potential for advanced brain cancer treatment with promising therapeutic outcomes.
Collapse
Affiliation(s)
- Masoud Farshbaf
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Mojarad-Jabali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamidreza Motasadizadeh
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Low Energy Status under Methionine Restriction Is Essentially Independent of Proliferation or Cell Contact Inhibition. Cells 2022; 11:cells11030551. [PMID: 35159360 PMCID: PMC8833905 DOI: 10.3390/cells11030551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023] Open
Abstract
Nonlimited proliferation is one of the most striking features of neoplastic cells. The basis of cell division is the sufficient presence of mass (amino acids) and energy (ATP and NADH). A sophisticated intracellular network permanently measures the mass and energy levels. Thus, in vivo restrictions in the form of amino acid, protein, or caloric restrictions strongly affect absolute lifespan and age-associated diseases such as cancer. The induction of permanent low energy metabolism (LEM) is essential in this process. The murine cell line L929 responds to methionine restriction (MetR) for a short time period with LEM at the metabolic level defined by a characteristic fingerprint consisting of the molecules acetoacetate, creatine, spermidine, GSSG, UDP-glucose, pantothenate, and ATP. Here, we used mass spectrometry (LC/MS) to investigate the influence of proliferation and contact inhibition on the energy status of cells. Interestingly, the energy status was essentially independent of proliferation or contact inhibition. LC/MS analyses showed that in full medium, the cells maintain active and energetic metabolism for optional proliferation. In contrast, MetR induced LEM independently of proliferation or contact inhibition. These results are important for cell behaviour under MetR and for the optional application of restrictions in cancer therapy.
Collapse
|
17
|
Zhi W, Li S, Wan Y, Wu F, Hong L. Short-term starvation synergistically enhances cytotoxicity of Niraparib via Akt/mTOR signaling pathway in ovarian cancer therapy. Cancer Cell Int 2022; 22:18. [PMID: 35016681 PMCID: PMC8753877 DOI: 10.1186/s12935-022-02447-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/03/2022] [Indexed: 12/21/2022] Open
Abstract
Background Short-term starvation (STS) has gradually been confirmed as a treatment method that synergistically enhances the effect of chemotherapy on malignant tumours. In clinical applications, there are still some limitations of poly (ADP-ribose) polymerase inhibitors (PARPi), including understanding their effectiveness and side effects. Here, we sought to investigate the effect and mechanism of the combined use of STS and niraparib in the treatment of ovarian cancer. Methods In in vitro experiments, SKOV3 and A2780 ovarian cancer cells were treated with STS and niraparib alone or in combination. Cell viability was assessed with CCK-8, and cell cycle, apoptosis, DNA damage repair and autophagy were examined to explore the molecular mechanisms. Akt and mTOR inhibitors were used to examine any changes in DNA damage repair levels. Xenograft animal models were treated with STS and niraparib, and HE staining and immunohistochemistry were performed to examine the effects. Results The combined use of STS and niraparib inhibited cell proliferation and increased apoptosis more than niraparib application alone. In addition, compared with the niraparib group, the STS + niraparib group had increased G2/M arrest, DNA damage and autophagy, which indicated that STS pretreatment enhanced the cytotoxicity of niraparib. In animal experiments, STS did not affect the growth of transplanted tumours, but the combined treatment synergistically enhanced the cytotoxicity of niraparib. In in vivo experiments, STS did not affect the growth of transplanted tumours, but the combined treatment synergistically enhanced the cytotoxicity of niraparib and reduced the small intestinal side effects caused by niraparib chemotherapy. Conclusion STS pretreatment can synergistically enhance the cytotoxicity of niraparib. STS + niraparib is a potentially effective strategy in the maintenance therapy of ovarian cancer.
Collapse
Affiliation(s)
- Wang Zhi
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei Province, People's Republic of China
| | - Suting Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei Province, People's Republic of China
| | - Yuting Wan
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei Province, People's Republic of China
| | - Fuwen Wu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei Province, People's Republic of China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei Province, People's Republic of China.
| |
Collapse
|
18
|
Bhatia R, Holtan S, Jurdi NE, Prizment A, Blaes A. Do Cancer and Cancer Treatments Accelerate Aging? Curr Oncol Rep 2022; 24:1401-1412. [PMID: 35796942 PMCID: PMC9606015 DOI: 10.1007/s11912-022-01311-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW This review focuses on describing the mechanisms and clinical manifestations that underlie accelerated aging associated with cancer and its treatment. RECENT FINDINGS The direct and indirect effects of cancer and its treatment are associated with late occurrence of comorbidities that happen earlier or more frequently in cancer survivors compared to cancer-free individuals, otherwise known as accelerated aging. Use of senolytics and dietary and exercise interventions including prehabilitation, caloric restriction, and rehabilitation are currently under investigation to reverse or decelerate the aging process and will be covered in this review. Further research on how to decelerate or reverse aging changes associated with cancer and its treatment will be of paramount importance as the number of cancer survivors continues to grow.
Collapse
Affiliation(s)
- Roma Bhatia
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114 USA
| | - Shernan Holtan
- grid.17635.360000000419368657Division of Hematology, Oncology and Transplantation, University of Minnesota, 425 E River Pkwy, Minneapolis, MN 55455 USA
| | - Najla El Jurdi
- grid.17635.360000000419368657Division of Hematology, Oncology and Transplantation, University of Minnesota, 425 E River Pkwy, Minneapolis, MN 55455 USA
| | - Anna Prizment
- grid.17635.360000000419368657Division of Hematology, Oncology and Transplantation, University of Minnesota, 425 E River Pkwy, Minneapolis, MN 55455 USA
| | - Anne Blaes
- grid.17635.360000000419368657Division of Hematology, Oncology and Transplantation, University of Minnesota, 425 E River Pkwy, Minneapolis, MN 55455 USA
| |
Collapse
|
19
|
Wilson KA, Chamoli M, Hilsabeck TA, Pandey M, Bansal S, Chawla G, Kapahi P. Evaluating the beneficial effects of dietary restrictions: A framework for precision nutrigeroscience. Cell Metab 2021; 33:2142-2173. [PMID: 34555343 PMCID: PMC8845500 DOI: 10.1016/j.cmet.2021.08.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Dietary restriction (DR) has long been viewed as the most robust nongenetic means to extend lifespan and healthspan. Many aging-associated mechanisms are nutrient responsive, but despite the ubiquitous functions of these pathways, the benefits of DR often vary among individuals and even among tissues within an individual, challenging the aging research field. Furthermore, it is often assumed that lifespan interventions like DR will also extend healthspan, which is thus often ignored in aging studies. In this review, we provide an overview of DR as an intervention and discuss the mechanisms by which it affects lifespan and various healthspan measures. We also review studies that demonstrate exceptions to the standing paradigm of DR being beneficial, thus raising new questions that future studies must address. We detail critical factors for the proposed field of precision nutrigeroscience, which would utilize individualized treatments and predict outcomes using biomarkers based on genotype, sex, tissue, and age.
Collapse
Affiliation(s)
| | - Manish Chamoli
- The Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Tyler A Hilsabeck
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Manish Pandey
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, India
| | - Sakshi Bansal
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, India
| | - Geetanjali Chawla
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, India.
| | - Pankaj Kapahi
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
20
|
Daily caloric restriction limits tumor growth more effectively than caloric cycling regardless of dietary composition. Nat Commun 2021; 12:6201. [PMID: 34707136 PMCID: PMC8551193 DOI: 10.1038/s41467-021-26431-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer incidence increases with age and is a leading cause of death. Caloric restriction (CR) confers benefits on health and survival and delays cancer. However, due to CR's stringency, dietary alternatives offering the same cancer protection have become increasingly attractive. Short cycles of a plant-based diet designed to mimic fasting (FMD) are protective against tumorigenesis without the chronic restriction of calories. Yet, it is unclear whether the fasting time, level of dietary restriction, or nutrient composition is the primary driver behind cancer protection. Using a breast cancer model in mice, we compare the potency of daily CR to that of periodic caloric cycling on FMD or an isocaloric standard laboratory chow against primary tumor growth and metastatic burden. Here, we report that daily CR provides greater protection against tumor growth and metastasis to the lung, which may be in part due to the unique immune signature observed with daily CR.
Collapse
|
21
|
Curran SP, Lithgow GJ, Verdin E, P C. University of Southern California and buck institute nathan shock center: multidimensional models of aging. GeroScience 2021; 43:2119-2127. [PMID: 34269983 PMCID: PMC8599784 DOI: 10.1007/s11357-021-00416-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022] Open
Abstract
The USC-Buck Nathan Shock Center of Excellence in the Biology of Aging is a new and fully integrated multi-institutional center focused on training the next generation of geroscientists and providing access to cutting-edge geroscience technologies to investigators across the nation. The USC-Buck NSC is devoted to forging a deeper understanding of how and why aging processes cause disease in order to advance the translation of basic research on aging into effective preventions and therapies. Including more than 61 NIA-supported investigators, six NIA-funded research centers, four NIA T32s, and several additional aging research centers of excellence, the USC-Buck NSC constitutes one of the largest collections of leaders in geroscience research within the USA; the unique nature of the USC-Buck NSC research infrastructure ensures an integrated organization that is representative of the wide breadth of topics encompassed by the biology of aging field. By leveraging the 25-year-long relationship, current collaborations and joint administrational activities of the University of Southern California and the Buck Institute for Aging Research, the USC-Buck NSC aims to enhance and expand promising research in the biology of aging at both at the and to make a positive impact across California, the nation and throughout the world. Specialized cores provide services to all Shock Center members, as well as provide support for services to the community at large.
Collapse
Affiliation(s)
- Sean P Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, USA.
| | | | - Eric Verdin
- Buck Institute for Research On Aging, Novato, CA, USA
- UCSF Department of Medicine, San Francisco, CA, USA
| | - Cohen P
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, USA
| |
Collapse
|
22
|
Huang AW, Wei M, Caputo S, Wilson ML, Antoun J, Hsu WC. An Intermittent Fasting Mimicking Nutrition Bar Extends Physiologic Ketosis in Time Restricted Eating: A Randomized, Controlled, Parallel-Arm Study. Nutrients 2021; 13:1523. [PMID: 33946428 PMCID: PMC8147148 DOI: 10.3390/nu13051523] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/26/2022] Open
Abstract
There has been increasing interest in time-restricted eating to attain intermittent fasting's metabolic benefits. However, a more extended daily fast poses many challenges. This study was designed to evaluate the effects of a 200-calorie fasting-mimicking diet (FMD) energy bar formulated to prolong ketogenesis and mitigate fasting-associated side effects. A randomized, controlled study was conducted comparing the impact of consuming an FMD bar vs. continued water fast, after a 15-h overnight fast. Subjects in the FMD group showed a 3-h postprandial beta-hydroxybutyrate (BHB) level and 4-h postprandial BHB area under the curve (AUC0-4) that were non-inferior to those who continued with the water fast (p = 0.891 and p = 0.377, respectively). The postprandial glucose AUC0-4 in the FMD group was non-inferior to that in the water fast group (p = 0.899). A breakfast group served as a control, which confirmed that the instrument used in home glucose and ketone monitoring functioned as expected. The results indicate that FMD bar consumption does not interfere with the physiological ketogenesis associated with overnight fasting and could be used to facilitate the practice of time-restricted eating or intermittent fasting.
Collapse
Affiliation(s)
- Angie W. Huang
- L-Nutra, Inc., Plano, TX 75024, USA; (A.W.H.); (M.W.); (S.C.); (J.A.)
| | - Min Wei
- L-Nutra, Inc., Plano, TX 75024, USA; (A.W.H.); (M.W.); (S.C.); (J.A.)
| | - Sara Caputo
- L-Nutra, Inc., Plano, TX 75024, USA; (A.W.H.); (M.W.); (S.C.); (J.A.)
| | - Melissa L. Wilson
- Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA;
| | - Joseph Antoun
- L-Nutra, Inc., Plano, TX 75024, USA; (A.W.H.); (M.W.); (S.C.); (J.A.)
| | - William C. Hsu
- L-Nutra, Inc., Plano, TX 75024, USA; (A.W.H.); (M.W.); (S.C.); (J.A.)
| |
Collapse
|
23
|
Travers S, Litofsky NS. Daily Lifestyle Modifications to Improve Quality of Life and Survival in Glioblastoma: A Review. Brain Sci 2021; 11:brainsci11050533. [PMID: 33922443 PMCID: PMC8146925 DOI: 10.3390/brainsci11050533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Survival in glioblastoma remains poor despite advancements in standard-of-care treatment. Some patients wish to take a more active role in their cancer treatment by adopting daily lifestyle changes to improve their quality of life or overall survival. We review the available literature through PubMed and Google Scholar to identify laboratory animal studies, human studies, and ongoing clinical trials. We discuss which health habits patients adopt and which have the most promise in glioblastoma. While results of clinical trials available on these topics are limited, dietary restrictions, exercise, use of supplements and cannabis, and smoking cessation all show some benefit in the comprehensive treatment of glioblastoma. Marital status also has an impact on survival. Further clinical trials combining standard treatments with lifestyle modifications are necessary to quantify their survival advantages.
Collapse
|
24
|
Guida JL, Agurs-Collins T, Ahles TA, Campisi J, Dale W, Demark-Wahnefried W, Dietrich J, Fuldner R, Gallicchio L, Green PA, Hurria A, Janelsins MC, Jhappan C, Kirkland JL, Kohanski R, Longo V, Meydani S, Mohile S, Niedernhofer LJ, Nelson C, Perna F, Schadler K, Scott JM, Schrack JA, Tracy RP, van Deursen J, Ness KK. Strategies to Prevent or Remediate Cancer and Treatment-Related Aging. J Natl Cancer Inst 2021; 113:112-122. [PMID: 32348501 PMCID: PMC7850536 DOI: 10.1093/jnci/djaa060] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/20/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
Up to 85% of adult cancer survivors and 99% of adult survivors of childhood cancer live with an accumulation of chronic conditions, frailty, and/or cognitive impairments resulting from cancer and its treatment. Thus, survivors often show an accelerated development of multiple geriatric syndromes and need therapeutic interventions. To advance progress in this area, the National Cancer Institute convened the second of 2 think tanks under the auspices of the Cancer and Accelerated Aging: Advancing Research for Healthy Survivors initiative. Experts assembled to share evidence of promising strategies to prevent, slow, or reverse the aging consequences of cancer and its treatment. The meeting identified research and resource needs, including geroscience-guided clinical trials; comprehensive assessments of functional, cognitive, and psychosocial vulnerabilities to assess and predict age-related outcomes; preclinical and clinical research to determine the optimal dosing for behavioral (eg, diet, exercise) and pharmacologic (eg, senolytic) therapies; health-care delivery research to evaluate the efficacy of integrated cancer care delivery models; optimization of intervention implementation, delivery, and uptake; and patient and provider education on cancer and treatment-related late and long-term adverse effects. Addressing these needs will expand knowledge of aging-related consequences of cancer and cancer treatment and inform strategies to promote healthy aging of cancer survivors.
Collapse
Affiliation(s)
- Jennifer L Guida
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Tim A Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | | | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca Fuldner
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Gallicchio
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Paige A Green
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | | | - Michelle C Janelsins
- Department of Surgery and Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Chamelli Jhappan
- Division of Cancer Biology, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Ronald Kohanski
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Valter Longo
- University of Southern California, Los Angeles, California, USA
- IFOM Institute, Milan, Italy
| | - Simin Meydani
- Jean Mayer USDA Human Nutritional Research Center on Aging, Tufts University, Boston, MA, USA
| | - Supriya Mohile
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Christian Nelson
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Frank Perna
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Keri Schadler
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Russell P Tracy
- Departments of Pathology & Laboratory Medicine, and Biochemistry, Larner College of Medicine, University of Vermont, Colchester, VT, USA
| | | | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
25
|
Fasting and fasting-mimicking diets for chemotherapy augmentation. GeroScience 2021; 43:1201-1216. [PMID: 33410090 DOI: 10.1007/s11357-020-00317-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/14/2020] [Indexed: 10/22/2022] Open
Abstract
The increasingly older population in most developed countries will likely experience aging-related chronic diseases such as diabetes, metabolic syndrome, heart and lung diseases, osteoporosis, arthritis, dementia, and/or cancer. Genetic and environmental factors, but also lifestyle choices including physical activity and dietary habits, play essential roles in disease onset and progression. Sixty-five percent of Americans diagnosed with cancer now survive more than 5 years, making the need for informed lifestyle choices particularly important to successfully complete their treatment, increase the recovery from the cytotoxic therapy options, and improve cancer-free survival. This review will discuss the findings on the use of prolonged fasting, as well as fasting-mimicking diets to augment cancer treatment. Preclinical studies in rodents strongly support the implementation of these dietary interventions and a small number of clinical trials begin to provide encouraging results for cancer patients and cancer survivors.
Collapse
|
26
|
Deligiorgi MV, Liapi C, Trafalis DT. How Far Are We from Prescribing Fasting as Anticancer Medicine? Int J Mol Sci 2020; 21:ijms21239175. [PMID: 33271979 PMCID: PMC7730661 DOI: 10.3390/ijms21239175] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022] Open
Abstract
(1) Background: the present review provides a comprehensive and up-to date overview of the potential exploitation of fasting as an anticancer strategy. The rationale for this concept is that fasting elicits a differential stress response in the setting of unfavorable conditions, empowering the survival of normal cells, while killing cancer cells. (2) Methods: the present narrative review presents the basic aspects of the hormonal, molecular, and cellular response to fasting, focusing on the interrelationship of fasting with oxidative stress. It also presents nonclinical and clinical evidence concerning the implementation of fasting as adjuvant to chemotherapy, highlighting current challenges and future perspectives. (3) Results: there is ample nonclinical evidence indicating that fasting can mitigate the toxicity of chemotherapy and/or increase the efficacy of chemotherapy. The relevant clinical research is encouraging, albeit still in its infancy. The path forward for implementing fasting in oncology is a personalized approach, entailing counteraction of current challenges, including: (i) patient selection; (ii) fasting patterns; (iii) timeline of fasting and refeeding; (iv) validation of biomarkers for assessment of fasting; and (v) establishment of protocols for patients’ monitoring. (4) Conclusion: prescribing fasting as anticancer medicine may not be far away if large randomized clinical trials consolidate its safety and efficacy.
Collapse
|
27
|
Wenger KJ, Wagner M, Harter PN, Franz K, Bojunga J, Fokas E, Imhoff D, Rödel C, Rieger J, Hattingen E, Steinbach JP, Pilatus U, Voss M. Maintenance of Energy Homeostasis during Calorically Restricted Ketogenic Diet and Fasting-MR-Spectroscopic Insights from the ERGO2 Trial. Cancers (Basel) 2020; 12:cancers12123549. [PMID: 33261052 PMCID: PMC7760797 DOI: 10.3390/cancers12123549] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/25/2020] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The glioblastoma is a highly malignant brain tumor with very limited treatment options up to date. Metabolism of this tumor is highly dependent on glucose uptake. It is believed that glioblastoma cells cannot metabolize ketone bodies, which are found in the blood during periods of fasting or ketogenic dieting. According to this hypothesis, dieting could lead to cancer cell starvation. The ERGO2 (Ernaehrungsumstellung bei Patienten mit Rezidiv eines Glioblastoms) MR-spectroscopic imaging subtrial was designed to investigate tumor metabolism in patients randomized to calorically restricted ketogenic diet/intermittent fasting versus standard diet. The non-invasive investigation of tumor metabolism is of high clinical interest. Abstract Background: The ERGO2 (Ernaehrungsumstellung bei Patienten mit Rezidiv eines Glioblastoms) MR-spectroscopic imaging (MRSI) subtrial investigated metabolism in patients randomized to calorically restricted ketogenic diet/intermittent fasting (crKD-IF) versus standard diet (SD) in addition to re-irradiation (RT) for recurrent malignant glioma. Intracerebral concentrations of ketone bodies (KB), intracellular pH (pHi), and adenosine triphosphate (ATP) were non-invasively determined. Methods: 50 patients were randomized (1:1): Group A keeping a crKD-IF for nine days, and Group B a SD. RT was performed on day 4–8. Twenty-three patients received an extended MRSI-protocol (1H decoupled 31P MRSI with 3D chemical shift imaging (CSI) and 2D 1H point-resolved spectroscopy (PRESS)) at a 3T scanner at baseline and on day 6. Voxels were selected from the area of recurrent tumor and contralateral hemisphere. Spectra were analyzed with LCModel, adding simulated signals of 3-hydroxybutyrate (βOHB), acetone (Acn) and acetoacetate (AcAc) to the standard basis set. Results: Acn was the only reliably MRSI-detectable KB within tumor tissue and/or normal appearing white matter (NAWM). It was detected in 4/11 patients in Group A and in 0/8 patients in Group B. MRSI results showed no significant depletion of ATP in tumor tissue of patients at day 6 during crKD-IF, even though there were a significant difference in ketone serum levels between Group A and B at day 6 and a decline in fasting glucose in Group A from baseline to day 6. The tumor specific alkaline pHi was maintained. Conclusions: Our metabolic findings suggest that tumor cells maintain energy homeostasis even with reduced serum glucose levels and may generate additional ATP through other sources.
Collapse
Affiliation(s)
- Katharina J. Wenger
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (M.W.); (E.H.); (U.P.)
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Correspondence: ; Tel.: +49-69-6301-80407
| | - Marlies Wagner
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (M.W.); (E.H.); (U.P.)
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
| | - Patrick N. Harter
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Neurological Institute (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Kea Franz
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Jörg Bojunga
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Department of Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Emmanouil Fokas
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Department of Radiotherapy and Oncology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Detlef Imhoff
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Department of Radiotherapy and Oncology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Claus Rödel
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Department of Radiotherapy and Oncology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Johannes Rieger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany;
- Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (M.W.); (E.H.); (U.P.)
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
| | - Joachim P. Steinbach
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany;
| | - Ulrich Pilatus
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (M.W.); (E.H.); (U.P.)
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
| | - Martin Voss
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (P.N.H.); (K.F.); (E.F.); (D.I.); (C.R.); (J.P.S.); (M.V.)
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany;
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany;
| |
Collapse
|
28
|
ERGO2: A Prospective, Randomized Trial of Calorie-Restricted Ketogenic Diet and Fasting in Addition to Reirradiation for Malignant Glioma. Int J Radiat Oncol Biol Phys 2020; 108:987-995. [DOI: 10.1016/j.ijrobp.2020.06.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 01/07/2023]
|
29
|
Pignatta S, Cortesi M, Arienti C, Zanoni M, Cocchi C, Sarnelli A, Arpa D, Piccinini F, Tesei A. Effects of radiotherapy and short-term starvation combination on metastatic and non-tumor cell lines. DNA Repair (Amst) 2020; 95:102949. [PMID: 32890865 DOI: 10.1016/j.dnarep.2020.102949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/22/2020] [Accepted: 08/08/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Since its discovery in the late 19th century, radiotherapy has been one of the most important medical treatments in oncology. Recently, fasting or short-term starvation (STS) in cancer patients undergoing chemotherapy has been studied to determine its potential for enhancing the therapeutic index and for preventing side- effects, but no data are available in the radiotherapy setting. We thus decided to investigate the effects in vitro of STS in combination with radiotherapy in metastatic cancer cells and non-cancer cells. METHODS Cells were incubated in short-term starvation medium (STS medium, 0·5 g/L glucose + 1% FBS) or in control medium (CM medium, 1 g/L glucose + 10 % FBS) for 24 h and then treated with single high-dose radiation. A plexiglass custom-built phantom was used to irradiate cells. DNA damage was evaluated using alkaline comet assay and theCometAnalyser software. The cell surviving fraction was assessed by clonogenic assay. FINDING STS followed by single high-dose radiation significantly increased DNA damage in metastatic cancer cell lines but not in normal cells. Furthermore, STS reduced the surviving fraction of irradiated tumor cells, indicating a good radio-sensitizing effect on metastatic cell lines. This effect was not observed in non-tumor cells. INTERPRETATION Our results suggest that STS may alter cellular processes, enhancing the efficacy of radiotherapy in metastatic cancer cellsin vitro. Interestingly, STS has radioprotective effect on the survival of healthy cells.
Collapse
Affiliation(s)
- Sara Pignatta
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy.
| | - Michela Cortesi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - Chiara Arienti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - Claudia Cocchi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - Anna Sarnelli
- Medical Physics Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Donatella Arpa
- Radiotherapy Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Filippo Piccinini
- Scientific Directorate, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Anna Tesei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy.
| |
Collapse
|
30
|
Zorn S, Ehret J, Schäuble R, Rautenberg B, Ihorst G, Bertz H, Urbain P, Raynor A. Impact of modified short-term fasting and its combination with a fasting supportive diet during chemotherapy on the incidence and severity of chemotherapy-induced toxicities in cancer patients - a controlled cross-over pilot study. BMC Cancer 2020; 20:578. [PMID: 32571329 PMCID: PMC7310229 DOI: 10.1186/s12885-020-07041-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This pilot trial aimed to investigate whether modified short-term fasting (mSTF) reduces the incidence of chemotherapy-induced toxicities and whether an initial ketogenic diet (KD) as fasting supportive diet reduces fasting-related discomfort and improves the compliance. METHODS In this controlled cross-over trial, gynaecologic cancer patients undergoing chemotherapy with a minimum of 4 cycles fasted for 96 h during half of their chemotherapy cycles and consumed a normocaloric diet during the other chemotherapy cycles. The caloric intake during mSTF was restricted to 25% of each patient's daily requirement. In addition, half of the patients should eat a 6-day normocaloric KD prior to each mSTF period to investigate a KD's hunger-suppression effect. Chemotherapy-induced toxicities, fasting-related discomfort, body composition, quality of life, laboratory values, and compliance were assessed at each chemotherapy. RESULTS Thirty patients aged 30-74 years (median 54 years) completed the study. During mSTF the frequency and severity score of stomatitis [- 0.16 ± 0.06; 95% CI -0.28 - (- 0.03); P = 0.013], headaches [- 1.80 ± 0.55; 95% CI -2.89 - (- 0.71); P = 0.002], weakness [- 1.99 ± 0.87; 95% CI -3.72 - (- 0.26); P = 0.024] and the total toxicities' score were significantly reduced [- 10.36 ± 4.44; 95% CI -19.22 - (- 1.50); P = 0.023]. We also observed significantly fewer chemotherapy postponements post-mSTF, reflecting improved tolerance of chemotherapy [- 0.80 ± 0.37; 95% CI -1.53 - (- 0.06); P = 0.034]. A significant reduction in mean body weight by - 0.79 ± 1.47 kg during mSTF was not compensated and remained until study's conclusion (P < 0.005). On average, Insulin [- 169.4 ± 44.1; 95% CI -257.1 - (- 81.8); P < 0.001] and Insulin-like growth factor 1 levels [- 33.3 ± 5.4; 95% CI -44.1 - (- 22.5); P < 0.001] dropped significantly during fasting. The KD as a fasting supportive diet neither reduced fasting-related discomfort nor improved compliance of our fasting regimen. CONCLUSION MSTF is safe and feasible in gynaecologic cancer patients. Our results indicate that mSTF during chemotherapy can reduce chemotherapy-induced toxicities and enhance the tolerance of chemotherapy. Larger clinical trials are required to recommend mSTF for cancer patients. TRIAL REGISTRATION germanctr.de: DRKS00011610, registered 30 January, 2017.
Collapse
Affiliation(s)
- Stefanie Zorn
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Janine Ehret
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rebecca Schäuble
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Beate Rautenberg
- Department of Gynecology and Gynecologic Oncology, Medical Center - University of Freiburg, Faculty of Gynecology, University of Freiburg, Freiburg, Germany
| | - Gabriele Ihorst
- Clinical Trials Unit, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Bertz
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Paul Urbain
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna Raynor
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
31
|
Abstract
Despite great advances in treatment, cancer remains a leading cause of death worldwide. Diet can greatly impact health, while caloric restriction and fasting have putative benefits for disease prevention and longevity. Strong epidemiological associations exist between obesity and cancer, whereas healthy diets can reduce cancer risk. However, less is known about how diet might impact cancer once it has been diagnosed and particularly how diet can impact cancer treatment. In the present review, we discuss the links between obesity, diet, and cancer. We explore potential mechanisms by which diet can improve cancer outcomes, including through hormonal, metabolic, and immune/inflammatory effects, and present the limited clinical research that has been published in this arena. Though data are sparse, diet intervention may reduce toxicity, improve chemotherapy efficacy, and lower the risk of long-term complications in cancer patients. Thus, it is important that we understand and expand the science of this important but complex adjunctive cancer treatment strategy.
Collapse
Affiliation(s)
- Steven D Mittelman
- Division of Pediatric Endocrinology, University of California, Los Angeles (UCLA), Children's Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA;
| |
Collapse
|
32
|
Jordan S, Tung N, Casanova-Acebes M, Chang C, Cantoni C, Zhang D, Wirtz TH, Naik S, Rose SA, Brocker CN, Gainullina A, Hornburg D, Horng S, Maier BB, Cravedi P, LeRoith D, Gonzalez FJ, Meissner F, Ochando J, Rahman A, Chipuk JE, Artyomov MN, Frenette PS, Piccio L, Berres ML, Gallagher EJ, Merad M. Dietary Intake Regulates the Circulating Inflammatory Monocyte Pool. Cell 2020; 178:1102-1114.e17. [PMID: 31442403 DOI: 10.1016/j.cell.2019.07.050] [Citation(s) in RCA: 265] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/02/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023]
Abstract
Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.
Collapse
Affiliation(s)
- Stefan Jordan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA.
| | - Navpreet Tung
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Maria Casanova-Acebes
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Christie Chang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Claudia Cantoni
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Dachuan Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Department of Cell Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, The Bronx, NY 10461, USA
| | - Theresa H Wirtz
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Shruti Naik
- Department of Pathology, and Ronald O. Perelman Department of Dermatology, NYU School of Medicine, 240 East 38(th) Street, New York, NY 10016, USA
| | - Samuel A Rose
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Bethesda, MD 20892, USA
| | - Anastasiia Gainullina
- Department of Pathology & Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Computer Technologies Department, ITMO University, Kronverksky 49, Saint Petersburg, Russian Federation
| | - Daniel Hornburg
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Sam Horng
- Department of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Barbara B Maier
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Paolo Cravedi
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Bethesda, MD 20892, USA
| | - Felix Meissner
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Adeeb Rahman
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Maxim N Artyomov
- Department of Pathology & Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Department of Cell Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, The Bronx, NY 10461, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA; Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown NSW 2050, Australia
| | - Marie-Luise Berres
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
33
|
Sofi F. FASTING-MIMICKING DIET a clarion call for human nutrition research or an additional swan song for a commercial diet? Int J Food Sci Nutr 2020; 71:921-928. [PMID: 32223459 DOI: 10.1080/09637486.2020.1746959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Numerous studies have reported beneficial effects for fasting in cellular and animal models, but human studies have shown conflicting results. Recently, a new diet has been introduced both in the scientific literature and in the lay media, the so-called fasting-mimicking diet. It is mainly characterised by a diet period that imitates fasting (generally 4-5 consecutive days) consisting of low or very low-calorie intake with a low or very low contribution of carbohydrates and proteins and a high fat intake. This protocol has been tested in some experimental animal models that have studied different outcomes, and in two small clinical trials that have reported some alleged beneficial effects especially on cardio-metabolic risk parameters. However, these clinical trials suffer from many limitations that require attention. The purpose of the present paper is to review the experimental and clinical studies that have investigated this particular dietary approach and to critically discuss the results.
Collapse
Affiliation(s)
- Francesco Sofi
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.,Unit of Clinical Nutrition, Careggi University Hospital, Florence, Italy
| |
Collapse
|
34
|
Castejón M, Plaza A, Martinez-Romero J, Fernandez-Marcos PJ, de Cabo R, Diaz-Ruiz A. Energy Restriction and Colorectal Cancer: A Call for Additional Research. Nutrients 2020; 12:E114. [PMID: 31906264 PMCID: PMC7019819 DOI: 10.3390/nu12010114] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023] Open
Abstract
: Colorectal cancer has the second highest cancer-related mortality rate, with an estimated 881,000 deaths worldwide in 2018. The urgent need to reduce the incidence and mortality rate requires innovative strategies to improve prevention, early diagnosis, prognostic biomarkers, and treatment effectiveness. Caloric restriction (CR) is known as the most robust nutritional intervention that extends lifespan and delays the progression of age-related diseases, with remarkable results for cancer protection. Other forms of energy restriction, such as periodic fasting, intermittent fasting, or fasting-mimicking diets, with or without reduction of total calorie intake, recapitulate the effects of chronic CR and confer a wide range of beneficial effects towards health and survival, including anti-cancer properties. In this review, the known molecular, cellular, and organismal effects of energy restriction in oncology will be discussed. Energy-restriction-based strategies implemented in colorectal models and clinical trials will be also revised. While energy restriction constitutes a promising intervention for the prevention and treatment of several malignant neoplasms, further investigations are essential to dissect the interplay between fundamental aspects of energy intake, such as feeding patterns, fasting length, or diet composition, with all of them influencing health and disease or cancer effects. Currently, effectiveness, safety, and practicability of different forms of fasting to fight cancer, particularly colorectal cancer, should still be contemplated with caution.
Collapse
Affiliation(s)
- Maria Castejón
- Nutritional Interventions Group, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, E-28049 Madrid, Spain; (M.C.); (R.d.C.)
| | - Adrian Plaza
- Bioactive Products and Metabolic Syndrome Group-BIOPROMET, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, E-28049 Madrid, Spain; (A.P.); (P.J.F.-M.)
| | - Jorge Martinez-Romero
- Molecular Oncology and Nutritional Genomics of Cancer Group, Precision Nutrition and Cancer Program, Institute IMDEA Food (CEI, UAM/CSIC), Crta. de Canto Blanco nº 8, E-28049 Madrid, Spain;
| | - Pablo Jose Fernandez-Marcos
- Bioactive Products and Metabolic Syndrome Group-BIOPROMET, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, E-28049 Madrid, Spain; (A.P.); (P.J.F.-M.)
| | - Rafael de Cabo
- Nutritional Interventions Group, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, E-28049 Madrid, Spain; (M.C.); (R.d.C.)
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Alberto Diaz-Ruiz
- Nutritional Interventions Group, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, E-28049 Madrid, Spain; (M.C.); (R.d.C.)
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| |
Collapse
|
35
|
de Groot S, Pijl H, van der Hoeven JJM, Kroep JR. Effects of short-term fasting on cancer treatment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:209. [PMID: 31113478 PMCID: PMC6530042 DOI: 10.1186/s13046-019-1189-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/22/2019] [Indexed: 12/14/2022]
Abstract
Growing preclinical evidence shows that short-term fasting (STF) protects from toxicity while enhancing the efficacy of a variety of chemotherapeutic agents in the treatment of various tumour types. STF reinforces stress resistance of healthy cells, while tumor cells become even more sensitive to toxins, perhaps through shortage of nutrients to satisfy their needs in the context of high proliferation rates and/or loss of flexibility to respond to extreme circumstances. In humans, STF may be a feasible approach to enhance the efficacy and tolerability of chemotherapy. Clinical research evaluating the potential of STF is in its infancy. This review focuses on the molecular background, current knowledge and clinical trials evaluating the effects of STF in cancer treatment. Preliminary data show that STF is safe, but challenging in cancer patients receiving chemotherapy. Ongoing clinical trials need to unravel if STF can also diminish toxicity and increase efficacy of chemotherapeutic regimes in daily practice.
Collapse
Affiliation(s)
- Stefanie de Groot
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC, Leiden, The Netherlands
| | - Hanno Pijl
- Department of Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, The Netherlands
| | - Jacobus J M van der Hoeven
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC, Leiden, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC, Leiden, The Netherlands.
| |
Collapse
|
36
|
Klement RJ, Pazienza V. Impact of Different Types of Diet on Gut Microbiota Profiles and Cancer Prevention and Treatment. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E84. [PMID: 30934960 PMCID: PMC6524347 DOI: 10.3390/medicina55040084] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 03/26/2019] [Indexed: 01/06/2023]
Abstract
: Diet is frequently considered as a food regimen focused on weight loss, while it is actually the sum of food consumed by the organism. Western diets, modern lifestyle, sedentary behaviors, smoking habits, and drug consumption have led to a significant reduction of gut microbial diversity, which is linked to many non-communicable diseases (NCDs). The latter kill 40 million people each year, equivalent to more than 70% of all deaths globally. Among NCDs, tumors play a major role, being responsible for 29% of deaths from NCDs. A link between diet, microbiota, and cancer prevention and treatment has recently been unveiled, underlining the importance of a new food culture based on limiting dietary surplus and on preferring healthier foods. Here, we review the effects of some of the most popular "cancer-specific" diets on microbiota composition and their potential impact on cancer prevention and treatment.
Collapse
Affiliation(s)
- Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Robert-Koch-Straße 10, 97422 Schweinfurt, Germany.
| | - Valerio Pazienza
- Gastroenterology Unit IRCCS "Casa Sollievo della Sofferenza", Hospital San Giovanni Rotondo, 71013 Foggia, Italy.
| |
Collapse
|
37
|
Caiola E, Falcetta F, Giordano S, Marabese M, Garassino MC, Broggini M, Pastorelli R, Brunelli L. Co-occurring KRAS mutation/LKB1 loss in non-small cell lung cancer cells results in enhanced metabolic activity susceptible to caloric restriction: an in vitro integrated multilevel approach. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:302. [PMID: 30514331 PMCID: PMC6280460 DOI: 10.1186/s13046-018-0954-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
Abstract
Background Non–small-cell lung cancer (NSCLC) is a heterogeneous disease, with multiple different oncogenic mutations. Approximately 25–30% of NSCLC patients present KRAS mutations, which confer poor prognosis and high risk of tumor recurrence. About half of NSCLCs with activating KRAS lesions also have deletions or inactivating mutations in the serine/threonine kinase 11 (LKB1) gene. Loss of LKB1 on a KRAS-mutant background may represent a significant source of heterogeneity contributing to poor response to therapy. Methods Here, we employed an integrated multilevel proteomics, metabolomics and functional in-vitro approach in NSCLC H1299 isogenic cells to define their metabolic state associated with the presence of different genetic background. Protein levels were obtained by label free and single reaction monitoring (SRM)-based proteomics. The metabolic state was studied coupling targeted and untargeted mass spectrometry (MS) strategy. In vitro metabolic dependencies were evaluated using 2-deoxy glucose (2-DG) treatment or glucose/glutamine nutrient limitation. Results Here we demonstrate that co-occurring KRAS mutation/LKB1 loss in NSCLC cells allowed efficient exploitation of glycolysis and oxidative phosphorylation, when compared to cells with each single oncologic genotype. The enhanced metabolic activity rendered the viability of cells with both genetic lesions susceptible towards nutrient limitation. Conclusions Co-occurrence of KRAS mutation and LKB1 loss in NSCLC cells induced an enhanced metabolic activity mirrored by a growth rate vulnerability under limited nutrient conditions relative to cells with the single oncogenetic lesions. Our results hint at the possibility that energy stress induced by calorie restriction regimens may sensitize NSCLCs with these co-occurring lesions to cytotoxic chemotherapy. Electronic supplementary material The online version of this article (10.1186/s13046-018-0954-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisa Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca Falcetta
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Silvia Giordano
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marina C Garassino
- Thoracic Oncology, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Laura Brunelli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy.
| |
Collapse
|
38
|
Abstract
The vulnerability of cancer cells to nutrient deprivation and their dependency on specific metabolites are emerging hallmarks of cancer. Fasting or fasting-mimicking diets (FMDs) lead to wide alterations in growth factors and in metabolite levels, generating environments that can reduce the capability of cancer cells to adapt and survive and thus improving the effects of cancer therapies. In addition, fasting or FMDs increase resistance to chemotherapy in normal but not cancer cells and promote regeneration in normal tissues, which could help prevent detrimental and potentially life-threatening side effects of treatments. While fasting is hardly tolerated by patients, both animal and clinical studies show that cycles of low-calorie FMDs are feasible and overall safe. Several clinical trials evaluating the effect of fasting or FMDs on treatment-emergent adverse events and on efficacy outcomes are ongoing. We propose that the combination of FMDs with chemotherapy, immunotherapy or other treatments represents a potentially promising strategy to increase treatment efficacy, prevent resistance acquisition and reduce side effects.
Collapse
Affiliation(s)
- Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | | | - Valter D Longo
- IFOM, FIRC Institute of Molecular Oncology, Milano, Italy.
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
39
|
Schübel R, Nattenmüller J, Sookthai D, Nonnenmacher T, Graf ME, Riedl L, Schlett CL, von Stackelberg O, Johnson T, Nabers D, Kirsten R, Kratz M, Kauczor HU, Ulrich CM, Kaaks R, Kühn T. Effects of intermittent and continuous calorie restriction on body weight and metabolism over 50 wk: a randomized controlled trial. Am J Clin Nutr 2018; 108:933-945. [PMID: 30475957 PMCID: PMC6915821 DOI: 10.1093/ajcn/nqy196] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 07/30/2018] [Indexed: 12/15/2022] Open
Abstract
Background Although preliminary evidence suggests that intermittent calorie restriction (ICR) exerts stronger effects on metabolic parameters, which may link obesity and major chronic diseases, compared with continuous calorie restriction (CCR), there is a lack of well-powered intervention studies. Objective We conducted a randomized controlled trial to test whether ICR, operationalized as the "5:2 diet," has stronger effects on adipose tissue gene expression, anthropometric and body composition measures, and circulating metabolic biomarkers than CCR and a control regimen. Design One hundred and fifty overweight and obese nonsmokers [body mass index (kg/m2) ≥25 to <40, 50% women], aged 35-65 y, were randomly assigned to an ICR group (5 d without energy restriction and 2 d with 75% energy deficit, net weekly energy deficit ∼20%), a CCR group (daily energy deficit ∼20%), or a control group (no advice to restrict energy) and participated in a 12-wk intervention phase, a 12-wk maintenance phase, and a 26-wk follow-up phase. Results Loge relative weight change over the intervention phase was -7.1% ± 0.7% (mean ± SEM) with ICR, -5.2% ± 0.6% with CCR, and -3.3% ± 0.6% with the control regimen (Poverall < 0.001, PICR vs. CCR = 0.053). Despite slightly greater weight loss with ICR than with CCR, there were no significant differences between the groups in the expression of 82 preselected genes in adipose tissue implicated in pathways linking obesity to chronic diseases. At the final follow-up assessment (week 50), weight loss was -5.2% ± 1.2% with ICR, -4.9% ± 1.1% with CCR, and -1.7% ± 0.8% with the control regimen (Poverall = 0.01, PICR vs. CCR = 0.89). These effects were paralleled by proportional changes in visceral and subcutaneous adipose tissue volumes. There were no significant differences between ICR and CCR regarding various circulating metabolic biomarkers. Conclusion Our results on the effects of the "5:2 diet" indicate that ICR may be equivalent but not superior to CCR for weight reduction and prevention of metabolic diseases. This trial was registered at clinicaltrials.gov as NCT02449148.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Diana Nabers
- Divisions of Cancer Epidemiology
- Medical and Biological Informatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Romy Kirsten
- Divisions of Cancer Epidemiology
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Biobank of the National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Mario Kratz
- Divisions of Cancer Epidemiology
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Cornelia M Ulrich
- Divisions of Cancer Epidemiology
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, UT
| | | | | |
Collapse
|
40
|
Employing proteomics to understand the effects of nutritional intervention in cancer treatment. Anal Bioanal Chem 2018; 410:6371-6386. [PMID: 29974151 DOI: 10.1007/s00216-018-1219-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/15/2018] [Accepted: 06/21/2018] [Indexed: 12/22/2022]
Abstract
Lifestyle optimizations are implementable changes that can have an impact on health and disease. Nutrition is a lifestyle optimization that has been shown to be of great importance in cancer initiation, progression, and metastasis. Dozens of clinical trials are currently in progress that focus on the nutritional modifications that cancer patients can make prior to and during medical care that increase the efficacy of treatment. In this review, we discuss various nutritional inventions for cancer patients and the analytical approaches to characterize the downstream molecular effects. We first begin by briefly explaining the many different forms of nutritional intervention currently being used in cancer treatment as well as their motivating biology. The forms of nutrient modulation described in this review include calorie restriction, the different practices of fasting, and carbohydrate restriction. The review then shifts to explain how proteomics is used to determine biomarkers of cancer and how it can be utilized in the future to determine the metabolic phenotype of a tumor, and inform physicians if nutritional intervention should be recommended for a cancer patient. Nutrigenomics aims to understand the relationship of nutrients and gene expression and can be used to understand the downstream molecular effects of nutrition restriction, partially through proteomic analysis. Proteomics is just beginning to be used as cancer diagnostic and predictive tools. However, these approaches have not been used to their full potential to understand nutritional intervention in cancer. Graphical abstract ᅟ.
Collapse
|
41
|
Hanjani NA, Vafa M. Protein Restriction, Epigenetic Diet, Intermittent Fasting as New Approaches for Preventing Age-associated Diseases. Int J Prev Med 2018; 9:58. [PMID: 30050669 PMCID: PMC6036773 DOI: 10.4103/ijpvm.ijpvm_397_16] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022] Open
Abstract
Data from epidemiological and experimental studies have shown that diet and eating patterns have a major role in the pathogenesis of many age-associated diseases. Since 1935, calorie restriction (CR) has been identified as one of the most effective nongenetic dietary interventions that can increase lifespan. It involves reducing calorie intake by about 20%–40% below ad libitum, without malnutrition. Restricting food intake has been observed to increase lifespan and prevent many age-associated diseases in rats, mice, and many other species. Understanding the metabolic, molecular, and cellular mechanisms involved in the anti-aging effects of CR can help us to find dietary interventions that can mimic its effects. Recently, different studies have shown that intermittent fasting, protein restriction, and an epigenetic diet can have similar effects to those of CR. These approaches were selected because it has been indicated that they act through a similar molecular pathway and also, are safe and effective in delaying or preventing diseases. In this review, we focus on the mechanistic pathway involved in CR. Then, we review the mimicking interventions through the mechanistic approach. For this purpose, we reviewed both animal and human articles, mainly available through the PubMed online database. We then selected the most relevant full texts which are summarized in this article.
Collapse
Affiliation(s)
- Nazanin Asghari Hanjani
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Buono R, Longo VD. Starvation, Stress Resistance, and Cancer. Trends Endocrinol Metab 2018; 29:271-280. [PMID: 29463451 PMCID: PMC7477630 DOI: 10.1016/j.tem.2018.01.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 12/20/2022]
Abstract
Cancer cells are characterized by dysregulation in signal transduction and metabolic pathways leading to increased glucose uptake, altered mitochondrial function, and the evasion of antigrowth signals. Fasting and fasting-mimicking diets (FMDs) provide a particularly promising intervention to promote differential effects in normal and malignant cells. These effects are caused in part by the reduction in IGF-1, insulin, and glucose and the increase in IGFBP1 and ketone bodies, which generate conditions that force cancer cells to rely more on metabolites and factors that are limited in the blood, thus resulting in cell death. Here we discuss the cellular and animal experiments demonstrating the differential effects of fasting on normal and cancer cells and the mechanisms responsible for these effects.
Collapse
Affiliation(s)
- Roberta Buono
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Valter D Longo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Caccialanza R, Cereda E, De Lorenzo F, Farina G, Pedrazzoli P. To fast, or not to fast before chemotherapy, that is the question. BMC Cancer 2018; 18:337. [PMID: 29587670 PMCID: PMC5870384 DOI: 10.1186/s12885-018-4245-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/19/2018] [Indexed: 12/28/2022] Open
Abstract
Fasting in disease prevention and treatment has recently become a popular topic, particularly in the context of oncology. Unfortunately, the growing attention paid by the media has created a background of speculations and ambiguous messages. The attitude towards the role of fasting in cancer patients should be very cautious, as the risk of malnutrition/sarcopenia and disinformation may be associated with this approach. Whether the results obtained by fasting in the cellular and animal models can be transferred to cancer patients is still to be ascertained. At the moment, more preclinical studies are required to determine in which cancers, at which stage, and in what combinations fasting, fasting-mimicking diets or caloric restriction mimetics may prove effective. So, despite the “rumors” of marketing and media, nowadays fasting and calorie restriction around CT represent only a promising intuition, which requires proper efforts and time to be validated by evidence-based clinical data.
Collapse
Affiliation(s)
- Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100, Pavia, Italy.
| | - Emanuele Cereda
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100, Pavia, Italy
| | | | - Gabriella Farina
- Department of Oncology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Paolo Pedrazzoli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
44
|
Yin J, Ren W, Huang X, Li T, Yin Y. Protein restriction and cancer. Biochim Biophys Acta Rev Cancer 2018; 1869:256-262. [PMID: 29596961 DOI: 10.1016/j.bbcan.2018.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/02/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
Protein restriction without malnutrition is currently an effective nutritional intervention known to prevent diseases and promote health span from yeast to human. Recently, low protein diets are reported to be associated with lowered cancer incidence and mortality risk of cancers in human. In murine models, protein restriction inhibits tumor growth via mTOR signaling pathway. IGF-1, amino acid metabolic programing, FGF21, and autophagy may also serve as potential mechanisms of protein restriction mediated cancer prevention. Together, dietary intervention aimed at reducing protein intake can be beneficial and has the potential to be widely adopted and effective in preventing and treating cancers.
Collapse
Affiliation(s)
- Jie Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, PR China; Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Xingguo Huang
- Department of Animal science, Hunan Agriculture University, Changsha, PR China; Hunan Co-Innovation Center of Animal Production Safety, Changsha, PR China
| | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, PR China; Hunan Co-Innovation Center of Animal Production Safety, Changsha, PR China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, PR China; Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, PR China.
| |
Collapse
|
45
|
Wei M, Brandhorst S, Shelehchi M, Mirzaei H, Cheng CW, Budniak J, Groshen S, Mack WJ, Guen E, Di Biase S, Cohen P, Morgan TE, Dorff T, Hong K, Michalsen A, Laviano A, Longo VD. Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci Transl Med 2017; 9:9/377/eaai8700. [PMID: 28202779 DOI: 10.1126/scitranslmed.aai8700] [Citation(s) in RCA: 340] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/23/2016] [Accepted: 12/20/2016] [Indexed: 12/28/2022]
Abstract
Calorie restriction or changes in dietary composition can enhance healthy aging, but the inability of most subjects to adhere to chronic and extreme diets, as well as potentially adverse effects, limits their application. We randomized 100 generally healthy participants from the United States into two study arms and tested the effects of a fasting-mimicking diet (FMD)-low in calories, sugars, and protein but high in unsaturated fats-on markers/risk factors associated with aging and age-related diseases. We compared subjects who followed 3 months of an unrestricted diet to subjects who consumed the FMD for 5 consecutive days per month for 3 months. Three FMD cycles reduced body weight, trunk, and total body fat; lowered blood pressure; and decreased insulin-like growth factor 1 (IGF-1). No serious adverse effects were reported. After 3 months, control diet subjects were crossed over to the FMD program, resulting in a total of 71 subjects completing three FMD cycles. A post hoc analysis of subjects from both FMD arms showed that body mass index, blood pressure, fasting glucose, IGF-1, triglycerides, total and low-density lipoprotein cholesterol, and C-reactive protein were more beneficially affected in participants at risk for disease than in subjects who were not at risk. Thus, cycles of a 5-day FMD are safe, feasible, and effective in reducing markers/risk factors for aging and age-related diseases. Larger studies in patients with diagnosed diseases or selected on the basis of risk factors are warranted to confirm the effect of the FMD on disease prevention and treatment.
Collapse
Affiliation(s)
- Min Wei
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Sebastian Brandhorst
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Mahshid Shelehchi
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Hamed Mirzaei
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Chia Wei Cheng
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Julia Budniak
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Susan Groshen
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Esra Guen
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Stefano Di Biase
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Pinchas Cohen
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Todd E Morgan
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Tanya Dorff
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kurt Hong
- Department of Internal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Andreas Michalsen
- Department of Internal and Complementary Medicine, Charité University Medical Center, 10117 Berlin, Germany
| | | | - Valter D Longo
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA. .,FIRC Institute of Molecular Oncology, Italian Foundation for Cancer Research Institute of Molecular Oncology, 20139 Milan, Italy
| |
Collapse
|
46
|
Senichkin VV, Kopeina GS, Prokhorova EA, Zamaraev AV, Lavrik IN, Zhivotovsky B. Modulation of Mcl-1 transcription by serum deprivation sensitizes cancer cells to cisplatin. Biochim Biophys Acta Gen Subj 2017; 1862:557-566. [PMID: 29203282 DOI: 10.1016/j.bbagen.2017.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/19/2017] [Accepted: 11/29/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND The development of approaches that increase therapeutic effects of anti-cancer drugs is one of the most important tasks of oncology. Caloric restriction in vivo or serum deprivation (SD) in vitro has been shown to be an effective tool for sensitizing cancer cells to chemotherapeutic drugs. However, the detailed mechanisms underlying the enhancement of apoptosis in cancer cells by SD remain to be elucidated. METHODS Flow cytometry, caspase activity assay and western blotting were used for cell death rate evaluation. Western blotting, gel-filtration, siRNA approach and qRT-PCR were used to elucidate the mechanism underlying cell death potentiation upon SD. RESULTS We demonstrated that SD sensitizes cancer cells to treatment with chemotherapeutic agent cisplatin. This effect is independent on activation of caspases-2 and -8, apical caspases triggering apoptosis in response to genotoxic stress. SD potentiates cell death via downregulation of the anti-apoptotic protein Mcl-1. In fact, SD reduces the Mcl-1 mRNA level, which consequently decreases the Mcl-1 protein level and renders cells more susceptible to apoptosis induction via the formation of apoptosome. CONCLUSIONS Mcl-1 protein is an important regulator of sensitivity of cancer cells to apoptotic stimuli upon SD. GENERAL SIGNIFICANCE This study identifies Mcl-1 as a new target for the sensitization of human cancer cells to cell death by SD, which is of great significance for the development of efficient anti-cancer therapies.
Collapse
Affiliation(s)
| | - Gelina S Kopeina
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia
| | | | - Alexey V Zamaraev
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia
| | - Inna N Lavrik
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia; Department of Translational Inflammation, Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Boris Zhivotovsky
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia; Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
47
|
Protective effects of short-term dietary restriction in surgical stress and chemotherapy. Ageing Res Rev 2017; 39:68-77. [PMID: 28216454 DOI: 10.1016/j.arr.2017.02.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 02/09/2017] [Accepted: 02/14/2017] [Indexed: 01/09/2023]
Abstract
Reduced caloric intake including fasting, as well as the dietary composition or the timing of food intake, impact longevity, likely through a modification in the onset or the severity of chronic aging-related diseases such as cancer. As with pre- and post-operative dietary recommendations, evidence-based nutritional advice from healthcare professionals during and after cancer treatment is often vague or conflicting. We hypothesize that preventive dietary recommendations can help in the context of both chronic cancer treatment efficacy and the avoidance of development of secondary malignancies, as well as in the context of protection from the acute stress of surgery. In this perspective review, we will discuss the latest findings on the potential role of short-term dietary restriction in cancer treatment and improvement of surgical outcome.
Collapse
|
48
|
Pietrocola F, Bravo-San Pedro JM, Galluzzi L, Kroemer G. Autophagy in natural and therapy-driven anticancer immunosurveillance. Autophagy 2017; 13:2163-2170. [PMID: 28598229 DOI: 10.1080/15548627.2017.1310356] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Autophagy is primordial for the maintenance of metabolic and genetic homeostasis in all eukaryotic organisms. Owing to its cell-intrinsic effects, autophagy robustly inhibits malignant transformation, yet can support the progression of established neoplasms as well as their resistance to conventional treatments. The notion that autophagy inhibition sensitizes neoplastic cells to chemotherapy and radiation therapy rivals with the capacity of autophagy to contribute to natural and therapy-driven anticancer immunosurveillance via a multitude of mechanisms. Indeed, autophagy ensures an optimal release of immunostimulatory signals by dying cancer cells and hence boosts their capacity to initiate an immune response. Moreover, autophagy is important for the activity of several components of the immune system involved in tumor recognition and elimination, including antigen-presenting cells and CD8+ cytotoxic T lymphocytes. In this review, we discuss how cancer cells disable autophagy to bypass immune control and how strategies aiming to enhance autophagy can be envisaged to improve the efficacy of immunogenic cancer therapies.
Collapse
Affiliation(s)
- Federico Pietrocola
- a Equipe 11 labellisée par la Ligue contre le Cancer , Centre de Recherche des Cordeliers , Paris , France.,b INSERM , U1138 , Paris , France.,c Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris , France.,d Université Pierre et Marie Curie/Paris VI , Paris , France.,e Metabolomics and Cell Biology Platforms , Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - José Manuel Bravo-San Pedro
- a Equipe 11 labellisée par la Ligue contre le Cancer , Centre de Recherche des Cordeliers , Paris , France.,b INSERM , U1138 , Paris , France.,c Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris , France.,d Université Pierre et Marie Curie/Paris VI , Paris , France.,e Metabolomics and Cell Biology Platforms , Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Lorenzo Galluzzi
- c Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris , France.,f Department of Radiation Oncology , Weill Cornell Medical College , New York , NY , USA.,g Sandra and Edward Meyer Cancer Center , New York, NY , USA
| | - Guido Kroemer
- a Equipe 11 labellisée par la Ligue contre le Cancer , Centre de Recherche des Cordeliers , Paris , France.,b INSERM , U1138 , Paris , France.,c Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris , France.,d Université Pierre et Marie Curie/Paris VI , Paris , France.,e Metabolomics and Cell Biology Platforms , Gustave Roussy Comprehensive Cancer Institute , Villejuif , France.,h Pôle de Biologie , Hopitâl Européen George Pompidou , AP-HP , Paris , France.,i Department of Women's and Children's Health , Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
49
|
Abstract
Cancer is the second leading cause of death in the USA and among the leading major diseases in the world. It is anticipated to continue to increase because of the growth of the aging population and prevalence of risk factors such as obesity, smoking, and/or poor dietary habits. Cancer treatment has remained relatively similar during the past 30 years with chemotherapy and/or radiotherapy in combination with surgery remaining the standard therapies although novel therapies are slowly replacing or complementing the standard ones. According to the American Cancer Society, the dietary recommendation for cancer patients receiving chemotherapy is to increase calorie and protein intake. In addition, there are no clear guidelines on the type of nutrition that could have a major impact on cancer incidence. Yet, various forms of reduced caloric intake such as calorie restriction (CR) or fasting demonstrate a wide range of beneficial effects able to help prevent malignancies and increase the efficacy of cancer therapies. Whereas chronic CR provides both beneficial and detrimental effects as well as major compliance challenges, periodic fasting (PF), fasting-mimicking diets (FMDs), and dietary restriction (DR) without a reduction in calories are emerging as interventions with the potential to be widely used to prevent and treat cancer. Here, we review preclinical and preliminary clinical studies on dietary restriction and fasting and their role in inducing cellular protection and chemotherapy resistance.
Collapse
|
50
|
Vernieri C, Casola S, Foiani M, Pietrantonio F, de Braud F, Longo V. Targeting Cancer Metabolism: Dietary and Pharmacologic Interventions. Cancer Discov 2016; 6:1315-1333. [PMID: 27872127 DOI: 10.1158/2159-8290.cd-16-0615] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022]
Abstract
Most tumors display oncogene-driven reprogramming of several metabolic pathways, which are crucial to sustain their growth and proliferation. In recent years, both dietary and pharmacologic approaches that target deregulated tumor metabolism are beginning to be considered for clinical applications. Dietary interventions exploit the ability of nutrient-restricted conditions to exert broad biological effects, protecting normal cells, organs, and systems, while sensitizing a wide variety of cancer cells to cytotoxic therapies. On the other hand, drugs targeting enzymes or metabolites of crucial metabolic pathways can be highly specific and effective, but must be matched with a responsive tumor, which might rapidly adapt. In this review, we illustrate how dietary and pharmacologic therapies differ in their effect on tumor growth, proliferation, and metabolism and discuss the available preclinical and clinical evidence in favor of or against each of them. We also indicate, when appropriate, how to optimize future investigations on metabolic therapies on the basis of tumor- and patient-related characteristics. SIGNIFICANCE To our knowledge, this is the first review article that comprehensively analyzes the preclinical and preliminary clinical experimental foundations of both dietary and pharmacologic metabolic interventions in cancer therapy. Among several promising therapies, we propose treatment personalization on the basis of tumor genetics, tumor metabolism, and patient systemic metabolism.Cancer Discov; 6(12); 1315-33. ©2016 AACR.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy. .,Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - Stefano Casola
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - Marco Foiani
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy.,Universita' degli Studi di Milano, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Universita' degli Studi di Milano, Milan, Italy
| | - Valter Longo
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy. .,Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, California
| |
Collapse
|