Buscar en
Clinics
Toda la web
Inicio Clinics Autophagy and intermittent fasting: the connection for cancer therapy?
Journal Information
Vol. 73. Issue S1.
(January 2018)
Share
Share
Download PDF
More article options
Visits
5296
Vol. 73. Issue S1.
(January 2018)
REVIEW ARTICLE
Open Access
Autophagy and intermittent fasting: the connection for cancer therapy?
Visits
5296
Fernanda AntunesI, Adolfo Garcia ErustesI, Angélica Jardim CostaI, Ana Carolina NascimentoI, Claudia BincolettoI, Rodrigo Portes UreshinoII, Gustavo José Silva PereiraI, Soraya Soubhi SmailiI,
Corresponding author
soraya.smaili23@gmail.com

Corresponding author
I Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (EPM-UNIFESP), Sao Paulo, SP, BR
II Departamento de Ciencias Biologicas, Universidade Federal de Sao Paulo, Diadema, SP, BR
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Special issue
This article is part of special issue:
Vol. 73. Issue S1
More info

Cancer is a leading cause of death worldwide, and its incidence is continually increasing. Although anticancer therapy has improved significantly, it still has limited efficacy for tumor eradication and is highly toxic to healthy cells. Thus, novel therapeutic strategies to improve chemotherapy, radiotherapy and targeted therapy are an important goal in cancer research. Macroautophagy (herein referred to as autophagy) is a conserved lysosomal degradation pathway for the intracellular recycling of macromolecules and clearance of damaged organelles and misfolded proteins to ensure cellular homeostasis. Dysfunctional autophagy contributes to many diseases, including cancer. Autophagy can suppress or promote tumors depending on the developmental stage and tumor type, and modulating autophagy for cancer treatment is an interesting therapeutic approach currently under intense investigation. Nutritional restriction is a promising protocol to modulate autophagy and enhance the efficacy of anticancer therapies while protecting normal cells. Here, the description and role of autophagy in tumorigenesis will be summarized. Moreover, the possibility of using fasting as an adjuvant therapy for cancer treatment, as well as the molecular mechanisms underlying this approach, will be presented.

KEYWORDS:
Apoptosis
Autophagy
Fasting
Cancer
Therapy
Full Text
Autophagy: definition and mechanisms

The 2016 Nobel Prize in Physiology or Medicine was awarded to Yoshinori Ohsumi for his initial elucidation of the morphological and molecular mechanisms of autophagy in the 1990s 1,2. Autophagy is an evolutionarily conserved lysosomal catabolic process by which cells degrade and recycle intracellular endogenous (damaged organelles, misfolded or mutant proteins and macromolecules) and exogenous (viruses and bacteria) components to maintain cellular homeostasis 3,4. The specificity of the cargo and the delivery route to lysosomes distinguishes the three major types of autophagy. Mircroautophagy involves the direct engulfment of cargo in endosomal/lysosomal membrane invaginations 5. Chaperone-mediated autophagy (CMA) recycles soluble proteins with an exposed amino acid motif (KFERQ) that is recognized by the heat shock protein hsc70; these proteins are internalized by binding to lysosomal receptors (LAMP-2A) 6. Macroautophagy (herein referred to as autophagy) is the best-characterized process; in this process, cytoplasmic constituents are engulfed within double-membrane vesicles called autophagosomes, which subsequently fuse with lysosomes to form autolysosomes, where the cargo are degraded or recycled 3,7. The degradation products include sugars, nucleosides/nucleotides, amino acids and fatty acids that can be redirected to new metabolic routes for cellular maintenance 8–10.

Autophagy occurs at basal levels under physiological conditions and can also be upregulated in response to stressful stimuli such as hypoxia, nutritional deprivation, DNA damage, and cytotoxic agents 11,12. The molecular machinery that mediates the autophagic process is evolutionarily conserved in higher eukaryotes and regulated by specific genes (ATG genes), which were initially characterized in yeast 13,14. Each stage is controlled by different protein complexes regulated by the activation or inactivation of several stress-responsive pathways, such as those involving mammalian target of rapamycin (mTOR—nutrient), AMP-activated protein kinase (AMPK—energy) and hypoxia-inducible factors (HIFs—stress) 3,15. Regarding initialization, the activation of the ULK1 complex (ULK1/2, Atg13, FIP200 and Atg101) signals for autophagosome nucleation under the control of the PI3K III complex (PI3KIII, Beclin-1, Atg14/Barkor, Vps15 and Ambra-1), whose activation induces PIP3 (phosphatidyl inositol 3 phosphate) production, which in turn recruits other Atg proteins to form the phagophore 16. Subsequently, two ubiquitin-like conjugation systems mediate the recruitment of ATG12–ATG5 and microtubule-associated protein light chain 3 (LC3) proteins to the phagophore, allowing its expansion and closure to form the mature autophagosome 17. This process leads to the conversion of the soluble protein LC3-I via conjugation to phosphatidylethanolamine to form an LC3-II membrane-associated form in the cytosol, specifically in the inner and outer membranes of the autophagosome 18,19. Furthermore, LC3-II can interact with adaptor proteins such as p62 (also known as sequestosome-1/SQSTM1), which directs cargo delivery to autophagosomes for further degradation in lysosomes, the final step of autophagy 20,21.

Throughout the past decade, autophagy has attracted considerable attention as a potential target of pharmacological agents or dietary interventions that inhibit or activate this process for several human disorders, including infections and inflammatory diseases 22, neurodegeneration 23, metabolic and cardiovascular diseases 24, obesity 25 and cancer 26,27.

Autophagy and cancer

The role of autophagy in cancer is complex, and its function may vary according to several biological factors, including tumor type, progression stage and genetic landscape, along with oncogene activation and tumor suppressor inactivation 26,28. Thus, autophagy can be related either to the prevention of tumorigenesis or to the enabling of cancer cell adaptation, proliferation, survival and metastasis 29,30. The initial indication that autophagy could have an important role in tumor suppression came from several studies exploring the essential autophagy gene BECN1, which encodes the Beclin-1 protein, in different cellular models. Liang et al. 31 demonstrated that BECN1 was frequently monoallelically deleted in ovarian, breast and testicular cancer. Moreover, mice harboring allelic loss of BECN1 had a partial autophagy deficiency and were prone to the development of hepatocarcinoma and lung tumors at an advanced age 32,33. However, BECN1 is located adjacent to the well-known tumor suppressor gene BRCA1, which is commonly deleted in hereditary breast cancer. These deletions are generally extensive and affect BRCA1 along with several other genes, including BECN1, suggesting that the deletion of BRCA1, not the deletion of BECN1, is the driver mutation in breast cancer 34. However, autophagy impairment due to a mosaic deletion of ATG5 induces benign liver tumors, demonstrating that different tissues have different responses to autophagy impairment 35. Furthermore, the activation of oncogenes (e.g., PI3KCA) and inactivation of tumor suppressors (e.g., PTEN and LKB1) are associated with autophagy inhibition and tumorigenesis 36. In general, studies from animal models note that the tumor suppressor function of autophagy is associated with cell protection from oxidative stress, DNA damage, inflammation and the accumulation of dysfunctional organelles. Collectively, these phenomena are important factors that could trigger genomic instabilities leading to tumor development 29,37,38. However, the loss of function of autophagy genes has not yet been identified and demonstrated in humans, raising doubts about the relevance of autophagy to tumor initiation in different types of cancer 26. In addition, the autophagic machinery is not a common target of somatic mutations, indicating that autophagy may have a fundamental role in the survival and progression of tumor cells 39.

Once the tumor is established, the main function of autophagy is to provide a means to cope with cellular stressors, including hypoxia, nutritional and growth factor deprivation and damaging stimuli, thus allowing tumor adaptation, proliferation, survival and dissemination 40. Autophagy, by degrading macromolecules and defective organelles, supplies metabolites and upregulates mitochondrial function, supporting tumor cell viability even in constantly stressful environments 11,29. Studies have demonstrated that autophagy increases in hypoxic regions of solid tumors, favoring cell survival. The inhibition of autophagy leads to an intense induction of cell death in these regions 41,42. Moreover, tumors frequently have mutations or deletions in the tumor suppressor protein p53, which also favors autophagy induction to recycle intracellular components for tumor growth 43. Although the basal autophagy rate is generally low in normal cells under physiological conditions, some tumors show a high level of basal autophagy, reinforcing the prosurvival role of autophagy in cancer 40,44. RAS-transformed cancer cells undergo autophagy upregulation to supply metabolic needs and maintain functional mitochondria, which in turn favors tumor establishment 45–47. Autophagy also has a supportive role in metastasis by interfering with epithelial-mesenchymal transition constituents to favor tumor cell dissemination 30. Finally, studies have demonstrated that autophagy is commonly induced as a survival mechanism against antitumor treatments, such as chemotherapy, radiotherapy and targeted therapy, contributing to treatment resistance 48,49.

Autophagy and cancer therapeutics

Because autophagy can inhibit tumor development or favor tumor growth, progression, invasion and treatment resistance, researchers proposed that autophagy modulation could be a new therapeutic strategy in the treatment of some malignancies 28,49,50.

Recently, we published a review on autophagy and cancer, suggesting that some challenges, such as the incomplete understanding of the relationship between autophagy, tumor resistance, and cell death, as well as the identification of new druggable targets, need to be overcome with the aim of pharmacologically modulating autophagy for cancer treatment 51. Some of these suggestions are based on the current literature and on previous studies published by our group demonstrating that combining different agents such as selumetinib and cytarabine with autophagy inhibitors (bafilomycin A1, chloroquine or 3-methyladenine) enhanced the activity of selumetinib and cytarabine against colorectal cancer cells 52 and leukemia cells 53, respectively. Autophagy was also observed in melanoma cells under treatment with palladium complex drugs 54, indicating the importance of investigating the relationship between autophagy and apoptosis during new drug development. Additionally, other studies demonstrated that inhibiting autophagy by chloroquine in combination with sorafenib in an in vitro model of glioblastoma 55 and in combination with temozolomide in melanoma patients augmented antitumor treatment efficacy 56. The inhibition of autophagy was also demonstrated to potentiate the response to radiotherapy in ovarian 57 and esophageal cancer 58. The efficacy of autophagy in favoring cell death has been demonstrated in many other cancer models, such as breast cancer, leukemia, prostate cancer, and myeloma 48,49. However, to date, clinical trials have not demonstrated that autophagy inhibition associated with anticancer therapy provided reliable therapeutic benefits to patients 59. Currently, protocols targeting autophagy induction instead of autophagy blockade are under intense investigation in oncology 28,50,60. Nevertheless, no drug currently licensed by any regulatory agency was developed for autophagy modulation, although several approved agents indeed modulate autophagy to some extent 61,62.

How does dietary restriction modulate autophagy and cancer therapy?

In preclinical studies, dietary restriction (DR) has been shown to extend the lifespan and reduce the development of age-related diseases such as diabetes, cancer, and neurodegenerative and cardiovascular diseases 63. DR promotes metabolic and cellular changes in organisms from prokaryotes to humans that allow adaptation to periods of limited nutrient availability 64. The main changes include decreased blood glucose levels and growth factor signaling and the activation of stress resistance pathways affecting cell growth, energy metabolism, and protection against oxidative stress, inflammation and cell death 64,65. Nutrient starvation also activates autophagy in most cultured cells and organs, such as the liver and muscle, as an adaptive mechanism to stressful conditions 11,66.

Studies demonstrate that dietary interventions can reduce tumor incidence and potentiate the effectiveness of chemo- and radiotherapy in different tumor models, highlighting dietary manipulation as a possible adjunct to standard cancer therapies 63,65. Among the many diet regimens that have been assessed, caloric restriction (CR) and fasting are the methods under intense investigation in oncology 63,65,67. CR is defined as a chronic reduction in the daily caloric intake by 20-40% without the incurrence of malnutrition and with the maintenance of meal frequency 68. In contrast, fasting is characterized by the complete deprivation of food but not water, with intervening periods of normal food intake. Based on the duration, fasting can be classified as (i) intermittent fasting (IF—e.g., alternate day fasting (≥16 hours) or 48 hours of fasting/week) or (ii) periodic fasting (PF—e.g., a minimum of 3 days of fasting every 2 or more weeks) 65. In this article, we do not review CR studies that have been reviewed elsewhere 63,68,69; instead, we focus on studies using IF protocols as an adjuvant to cancer treatment in animals and humans.

Recently, studies in in vitro and in vivo models have shown that intermittent fasting improved the chemotherapeutic response to cisplatin, doxorubicin, cyclophosphamide 70, oxaliplatin 71, sorafenib 72, mitoxantrone 73, gemcitabine 74, etoposide 75, temozolomide 76 and tyrosine kinase inhibitors 77 in models of glioma, neuroblastoma, melanoma, fibrosarcoma and breast cancer, colon cancer, pancreatic cancer, hepatocellular cancer and lung cancer. IF has also been shown to improve the radiosensitivity of glioma 76 and breast cancer 78 in mice. Interestingly, fasting in combination with cytotoxic agents elicited differential responses in normal and cancer cells, a phenomenon known as differential stress resistance (DSR). For DSR, normal cells prioritize maintenance pathways and inactivate growth factor signaling when nutrients are absent. In contrast, cancer cells, due to oncogene activation, do not inhibit stress resistance pathways, thus becoming vulnerable to cytotoxic treatment 70,75. IF, by reducing the circulating glucose levels, protected mice from doxorubicin toxicity and particularly promoted cardioprotection mediated in part by EGFR1-dependent transcriptional regulation of atrial natriuretic peptide and B-type natriuretic peptide in heart tissue 79. As demonstrated by Tinkum et al. 80, IF also facilitated DNA repair activation mechanisms and preserved small intestinal (SI) stem cell viability as well SI architecture and barrier function after exposure to high-dose etoposide, suggesting that fasting can be applied to reduce side effects and toxicity in patients undergoing chemotherapy.

Although the results of combining IF with anticancer drugs are encouraging, the molecular mechanisms are not completely clear. Lee et al. 81 demonstrated that IF (48-hour fasting) reduced the glucose and IGF-1 levels by 60% and 70%, respectively, in a breast cancer animal model. In a colon cancer model, IF inhibited tumor growth without causing permanent weight loss and decreased M2 polarization of tumor-associated macrophages in mice. In vitro data showed autophagy induction and CD73 downregulation, followed by a decrease in extracellular adenosine and the inhibition of M2 polarization due to the inactivation of JAK1/STAT3 82.

When IF cycles were combined with chemotherapy, tumor growth was slowed and overall survival was prolonged in breast cancer, melanoma and neuroblastoma animal models 70. The in vitro data showed that this therapeutic combination resulted in increased Akt and S6 kinase phosphorylation, caspase-3 cleavage and apoptosis induction in cancer cells but not in normal cells 70. Other studies demonstrated that the combination of IF and oxaliplatin also reduced tumor growth and glucose uptake in vivo and resulted in downregulated aerobic glycolysis followed by augmented oxidative phosphorylation, leading to increased oxidative stress, decreased ATP synthesis and cell death in colon cancer cell models 71. Furthermore, Our group also demonstrated that nutritional deprivation enhanced the sensitivity of both wild type and BRAFV600E human melanoma cells to cisplatin treatment followed by ROS production and mitochondrial perturbation leading to apoptosis without autophagy involvement in the cell death process 83. Pietrocola et al. 73 showed that IF improved the chemotherapeutic response to mitoxantrone and oxaliplatin in murine fibrosarcoma, reducing tumor growth in immunocompetent mice. This group also showed that the impairment of tumor growth was dependent on the cellular immune system as well as on autophagy; IF + chemotherapy could not impair tumor growth in either athymic nu/nu mice or tumor cells after autophagy deficiency was induced by Atg5 knockdown.

The combination of IF and tyrosine kinase inhibitors such as erlotinib, gefitinib, lapatinib, crizotinib and regorafenib promoted the sustained inhibition of the MAPK pathway, leading to antiproliferative effects in breast, colorectal and lung cancer cell models, as well as to the inhibition of tumor growth in an in vivo model of lung cancer 77. The combination of IF and the multi-tyrosine kinase inhibitor sorafenib exhibited an additive effect in inhibiting hepatocarcinoma cell proliferation and glucose uptake as well as downregulating the MAPK pathway and the gene expression of BIRC5, DKK1, TRIB3 and VEGF, which are commonly altered in hepatocarcinoma cells 72. In pancreatic cancer, fasting increased the uptake of gemcitabine due to enhanced levels of its transporter (hENT1), thus potentiating cell death. In a xenograft pancreatic cancer model, fasting cycles and gemcitabine treatment induced a reduction in tumor growth of more than 40% 74.

A small pilot study comprising 10 patients diagnosed with breast, prostate, esophageal or lung cancer in advanced stages suggested that periods of intermittent fasting before and after chemotherapy reduces the self-reported side effects of therapy, especially those associated with the gastrointestinal system, as well as weakness and fatigue. Additionally, no negative effect on the chemotherapy response or persistent weight loss was observed 84,85. In another clinical trial, the combination of IF and platinum-based chemotherapy promoted pathologic complete or partial radiographic responses in the majority of patients affected by different stages and types of tumors, such as ovarian, uterine, breast and urothelial cancer. A reduction in leukocyte DNA damage, in addition to decreased levels of circulating IGF-1, has also been reported 86. Both studies established the feasibility of IF in humans and suggested that combining IF with cytotoxic agents in the clinical context is safe and may be well-tolerated by patients, although this regimen may be psychologically uncomfortable for some individuals 84–87. Currently, other clinical trials involving IF combined with chemotherapy in cancer patients are underway; these trials are summarized in Table 1. The results of these trials will be essential for a better evaluation of the clinical potential and application of this new therapeutic strategy.

Table 1.

Completed and current clinical trials investigating the effects of fasting as adjunct therapy to anti-cancer treatment.

Cancer/Phase  Treatment  Outcome/Status  Reference 
Breast Cancer, Hormone-resistant Prostate Cancer, Recurrent Prostate Cancer  Chemotherapy + low-calorie diet  Currently recruiting participants  NCT01802346 
Advanced Metastatic Prostate Cancer  Chemotherapy + fasting and nutritional therapy  Currently recruiting participants  NCT02710721 
HER2 Negative Breast Cancer  Chemotherapy + fasting mimicking diet  Currently recruiting participants  NCT02126449 
Breast Cancer  Chemotherapy + short-term fasting (IF)  IF associated with chemotherapy was well tolerated, reduced hematological toxicity in HER2-negative BC patients and also induced a faster recovery of DNA damage in PBMCs (peripheral blood mononuclear cells)  NCT01304251 (96
Gynecological cancer disease (ovarian and breast cancer)  Chemotherapy + short-term fasting  Completed, no results reported  NCT01954836 
Breast cancer  Chemotherapy + short-term fasting  Completed, no results reported  NCT02379585 
Malignant Neoplasm  Short-term fasting prior to systemic chemotherapy  Active  NCT01175837 
Malignant Neoplasm  Chemotherapy + fasting  Completed, no results reported  NCT00757094 

Another novel pharmacological therapeutic strategy currently being investigated to treat cancer is the combination of caloric restriction mimetics (CRMs) with cytotoxic agents. CRMs are compounds that have different chemical structures and mimic the biochemical and functional effects of CR, such as the activation of AMPK and inhibition of mTOR leading to autophagy induction, the depletion of acetyl-CoA and ATP, and the reduced utilization of glucose, without eliciting the discomfort of CR 88. Several studies demonstrated the tumor-suppressive effects of CRM agents, for example, 2-deoxy-glucose 89, metformin 90,91, mTOR inhibitors 92, resveratrol 73,93, hydroxycitrate 73, spermidine 73,94 and natural compounds such as curcumin 95, in combination with antitumor treatments in different cancer models. The possible connections between fasting and anticancer therapy potentiation in tumor cells are summarized in Figure 1.

Figure 1.

Presumable molecular mechanisms induced by fasting and anticancer treatment to promote intracellular changes and autophagy induction in tumor cells. I) Fasting may oppose the Warburg effect (glucose breakdown by glycolysis even in the presence of oxygen), favoring oxidative phosphorylation in tumor cells and resulting in increased ROS production and reduced levels of lactate and possibly ATP. The increase in the ADP/ATP ratio can activate the AMPK pathway, leading to autophagy induction. Moreover, the sustained stressful environment can result in cell death induction. II) Several tumors harbor mutations that favor MAPK pathway hyperactivation, which enables tumor cell growth, survival and proliferation. Therapies targeting this pathway, as well as fasting, may result in the downregulation of this pathway alongside a reduction in AKT and mTOR activation, resulting in autophagy induction and cell death. III) Furthermore, fasting potentiates the detrimental effects of chemotherapy, such as DNA damage, thus activating the cell death machinery, deregulating pro- and antiapoptotic proteins, and inducing mitochondrial alterations and caspase activation, which in turn culminates in apoptosis.

(0.06MB).

In this review, we highlighted the concepts of autophagy, especially in relation to tumorigenesis, as well as the potential of autophagy as a therapeutic target in the treatment of different malignancies. We also pointed out the possibility of using dietary manipulation as an autophagy modulator as well as a cost-effective intervention to increase therapeutic response in the challenging oncologic arena. Furthermore, fasting may protect normal cells from the toxicity of anticancer agents, reducing side effects in patients and increasing the detrimental effects of chemotherapy, radiotherapy and targeted therapy on tumor cells. However, additional studies are required to better understand the molecular mechanisms evoked by fasting, aiming to identify the context in which fasting may be beneficial as an adjunct to cancer treatment. Moreover, further knowledge may also lead to the development of novel pharmacological protocols that replicate effects similar to those of fasting and are more suitable for different oncologic patients.

AUTHOR CONTRIBUTIONS

Antunes F contributed to the design of the study, wrote most of the study and edited the manuscript. Erustes AG, Costa AJ, Nascimento AC and Trindade CB wrote the manuscript. Ureshino RP, Pereira GJ and Smaili SS wrote, designed and coordinated the study and edited and reviewed the final version of the manuscript. All authors reviewed and approved the final version of the manuscript.

ACKNOWLEDGMENTS

The authors are grateful for the financial support given by the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) (08/11515-3 and 13/20073-2 by Smaili SS), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Coordenação de Aperfeiçoamento de Pessoal de Ensino Superior (CAPES).

REFERENCES
[1]
M Tsukada , Y Ohsumi .
Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae.
[2]
K Takeshige , M Baba , S Tsuboi , T Noda , Y Ohsumi .
Autophagy in yeast demonstrated with proteinase-deficient mutants and conditions for its induction.
[3]
L Galluzzi , EH Baehrecke , A Ballabio , P Boya , JM Bravo-San Pedro , F Cecconi , et al.
Molecular definitions of autophagy and related processes.
[4]
B Levine , DJ Klionsky .
Development by self-digestion: molecular mechanisms and biological functions of autophagy.
[5]
D Mijaljica , M Prescott , RJ Devenish .
Microautophagy in mammalian cells: revisiting a 40-year-old conundrum.
[6]
S Kaushik , AM Cuervo .
Chaperone-mediated autophagy: a unique way to enter the lysosome world.
[7]
N Mizushima , M Komatsu .
Autophagy: renovation of cells and tissues.
[8]
AC Kimmelman , E White .
Autophagy and Tumor Metabolism.
[9]
RA Saxton , DM Sabatini .
mTOR Signaling in Growth, Metabolism, and Disease.
[10]
C Settembre , R De Cegli , G Mansueto , PK Saha , F Vetrini , O Visvikis , et al.
TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop.
[11]
G Kroemer , G Marião , B Levine .
Autophagy and the integrated stress response.
[12]
J Kaur , J Debnath .
Autophagy at the crossroads of catabolism and anabolism.
Nat Rev Mol Cell Biol, 16 (2015), pp. 461-472
[13]
N Mizushima , T Yoshimori , Y Ohsumi .
The role of Atg proteins in autophagosome formation.
[14]
DJ Klionsky , JM Cregg , WA Dunn Jr , SD Emr , Y Sakai , IV Sandoval , et al.
A unified nomenclature for yeast autophagy-related genes.
[15]
M Antonioli , M Di Rienzo , M Piacentini , GM Fimia .
Emerging Mechanisms in Initiating and Terminating Autophagy.
[16]
MG Lin , JH Hurley .
Structure and function of the ULK1 complex in autophagy.
[17]
N Mizushima , T Noda , T Yoshimori , Y Tanaka , T Ishii , MD George , et al.
A protein conjugation system essential for autophagy.
[18]
Y Ichimura , T Kirisako , T Takao , Y Satomi , Y Shimonishi , N Ishihara , et al.
A ubiquitin-like system mediates protein lipidation.
[19]
Z Yang , DJ Klionsky .
Mammalian autophagy: core molecular machinery and signaling regulation.
[20]
S Pankiv , TH Clausen , T Lamark , A Brech , JA Bruun , H Outzen , et al.
p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy.
[21]
A Puissant , N Fenouille , P Auberger .
When autophagy meets cancer through p62/SQSTM1.
Am J Cancer Res, 2 (2012), pp. 397-413
[22]
K Cadwell .
Crosstalk between autophagy and inflammatory signalling pathways: balancing defence and homeostasis.
[23]
FM Menzies , A Fleming , A Caricasole , CF Bento , SP Andrews , A Ashkenazi , et al.
Autophagy and Neurodegeneration: Pathogenic Mechanisms and Therapeutic Opportunities.
[24]
JM Bravo-San Pedro , G Kroemer , L Galluzzi .
Autophagy and Mitophagy in Cardiovascular Disease.
[25]
VJ Lavallard , AJ Meijer , P Codogno , P Gual .
Autophagy, signaling and obesity.
[26]
R Amaravadi , AC Kimmelman , E White .
Recent insights into the function of autophagy in cancer.
[27]
L Galluzzi , F Pietrocola , JM Bravo-San Pedro , RK Amaravadi , EH Baehrecke , F Cecconi , et al.
Autophagy in malignant transformation and cancer progression.
[28]
JM Levy , CG Towers , A Thorburn .
Targeting autophagy in cancer.
[29]
E White .
Deconvoluting the context-dependent role for autophagy in cancer.
[30]
MN Sharifi , EE Mowers , LE Drake , C Collier , H Chen , M Zamora , et al.
Autophagy Promotes Focal Adhesion Disassembly and Cell Motility of Metastati Tumor Cells through the Direct Interaction of Paxillin with LC3.
[31]
XH Liang , S Jackson , M Seaman , K Brown , B Kempkes , H Hibshoosh , et al.
Induction of autophagy and inhibition of tumorigenesis by beclin 1.
[32]
X Qu , J Yu , G Bhagat , N Furuya , H Hibshoosh , A Troxel , J Rosen , et al.
Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene.
[33]
Z Yue , S Jin , C Yang , AJ Levine , N Heintz .
Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor.
[34]
SV Laddha , S Ganesan , CS Chan , E White .
Mutational landscape of the essential autophagy gene BECN1 in human cancers.
[35]
A Takamura , M Komatsu , T Hara , A Sakamoto , C Kishi , S Waguri , et al.
Autophagy-deficient mice develop multiple liver tumors.
[36]
C He , B Levine .
The Beclin 1 interactome.
[37]
R Mathew , S Kongara , B Beaudoin , CM Karp , K Bray , K Degenhardt , et al.
Autophagy suppresses tumor progression by limiting chromosomal instability.
[38]
R Mathew , E White .
Autophagy, stress, and cancer metabolism: what doesn’t kill you makes you stronger.
[39]
CB Lebovitz , AG Robertson , R Goya , SJ Jones , RD Morin , MA Marra , et al.
Cross-cancer profiling of molecular alterations within the human autophagy interaction network.
[40]
E White .
The role for autophagy in cancer.
[41]
K Degenhardt , R Mathew , B Beaudoin , K Bray , D Anderson , G Chen , et al.
Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis.
[42]
I Papandreou , AL Lim , K Laderoute , NC Denko .
Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L.
[43]
E Tasdemir , MC Maiuri , L Galluzzi , I Vitale , M Djavaheri-Mergny , M D’Amelio , et al.
Regulation of autophagy by cytoplasmic p53.
[44]
M Corazzari , F Rapino , F Ciccosanti , P Giglio , M Antonioli , B Conti , et al.
Oncogenic BRAF induces chronic ER stress condition resulting in increased basal autophagy and apoptotic resistance of cutaneous melanoma.
[45]
R Lock , CM Kenific , AM Leidal , E Salas , J Debnath .
Autophagy-dependent production of secreted factors facilitates oncogenic RAS-driven invasion.
[46]
R Lock , S Roy , CM Kenific , JS Su , E Salas , SM Ronen , et al.
Autophagy facilitates glycolysis during Ras-mediated oncogenic transformation.
[47]
JY Guo , HY Chen , R Mathew , J Fan , AM Strohecker , G Karsli-Uzunbas , et al.
Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis.
[48]
N Chen , V Karantza .
Autophagy as a therapeutic target in cancer.
[49]
L Galluzzi , JM Bravo-San Pedro , B Levine , DR Green , G Kroemer .
Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles.
[50]
S Fulda .
Autophagy in Cancer Therapy.
[51]
C Bincoletto , A Bechara , GJ Pereira , CP Santos , F Antunes , J Peixoto da-Silva , et al.
Interplay between apoptosis and autophagy, a challenging puzzle: new perspectives on antitumor chemotherapies.
[52]
S Grasso , GJ Pereira , C Palmeira-Dos-Santos , AK Calgarotto , I Martínez-Lacaci , JA Ferragut , et al.
Autophagy regulates Selumetinib (AZD6244) induced-apoptosis in colorectal cancer cells.
[53]
C Palmeira dos Santos , GJ Pereira , CM Barbosa , A Jurkiewicz , SS Smaili , C Bincoletto .
Comparative study of autophagy inhibition by 3MA and CQ on Cytarabine-induced death of leukaemia cells.
[54]
R Gigli , GJ Pereira , F Antunes , A Bechara , DM Garcia , DG Spindola , et al.
The biphosphinic paladacycle complex induces melanoma cell death through lysosomal-mitochondrial axis modulation and impaired autophagy.
[55]
X Liu , K Sun , H Wang , Y Dai .
Inhibition of Autophagy by Chloroquine Enhances the Antitumor Efficacy of Sorafenib in Glioblastoma.
[56]
R Rangwala , R Leone , YC Chang , LA Fecher , LM Schuchter , A Kramer , et al.
Phase I trial of hydroxychloroquine with dose-intense temozolomide in patients with advanced solid tumors and melanoma.
[57]
B Liang , D Kong , Y Liu , N Liang , M He , S Ma , et al.
Autophagy inhibition plays the synergetic killing roles with radiation in the multi-drug resistant SKVCR ovarian cancer cells.
[58]
Y Chen , X Li , L Guo , X Wu , C He , S Zhang , et al.
Combining radiation with autophagy inhibition enhances suppression of tumor growth and angiogenesis in esophageal cancer.
[59]
DA Gewirtz .
The Challenge of Developing Autophagy Inhibition as a Therapeutic Strategy.
[60]
L Galluzzi , JM Bravo-San Pedro , S Demaria , SC Formenti , G Kroemer .
Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy.
[61]
H Vakifahmetoglu-Norberg , HG Xia , J Yuan .
Pharmacologic agents targeting autophagy.
[62]
J Ha , J Kim .
Novel pharmacological modulators of autophagy: an updated patent review (2012-2015).
[63]
CH O’Flanagan , LA Smith , SB McDonell , SD Hursting .
When less may be more: calorie restriction and response to cancer therapy.
[64]
VD Longo , MP Mattson .
Fasting: molecular mechanisms and clinical applications.
[65]
MP Mattson , VD Longo , M Harvie .
Impact of intermittent fasting on health and disease processes.
[66]
N Mizushima , A Yamamoto , M Matsui , T Yoshimori , Y Ohsumi .
In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker.
[67]
BA Simone , CE Champ , AL Rosenberg , AC Berger , DA Monti , AP Dicker , et al.
Selectively starving cancer cells through dietary manipulation: methods and clinical implications.
[68]
JR Speakman , SE Mitchell .
Caloric restriction.
[69]
GS Kopeina , VV Senichkin , B Zhivotovsky .
Caloric restriction - A promising anti-cancer approach: From molecular mechanisms to clinical trials.
[70]
C Lee , L Raffaghello , S Brandhorst , FM Safdie , G Bianchi , A Martin-Montalvo , et al.
Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy.
[71]
G Bianchi , R Martella , S Ravera , C Marini , S Capitanio , A Orengo , et al.
Fasting induces anti-Warburg effect that increases respiration but reduces ATP-synthesis to promote apoptosis in colon cancer models.
[72]
O Lo Re , C Panebianco , S Porto , C Cervi , F Rappa , S Di Biase , et al.
Fasting inhibits hepatic stellate cells activation and potentiates anti-cancer activity of Sorafenib in hepatocellular cancer cells.
[73]
F Pietrocola , J Pol , E Vacchelli , S Rao , DP Enot , EE Baracco , et al.
Caloric Restriction Mimetics Enhance Anticancer Immunosurveillance.
[74]
M D’Aronzo , M Vinciguerra , T Mazza , C Panebianco , C Saracino , SP Pereira , et al.
Fasting cycles potentiate the efficacy of gemcitabine treatment in in vitro and in vivo pancreatic cancer models.
[75]
L Raffaghello , C Lee , FM Safdie , M Wei , F Madia , G Bianchi , et al.
Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy.
[76]
F Safdie , S Brandhorst , M Wei , W Wang , C Lee , S Hwang , et al.
Fasting enhances the response of glioma to chemo- and radiotherapy.
[77]
I Caffa , V D’Agostino , P Damonte , D Soncini , M Cea , F Monacelli , et al.
Fasting potentiates the anticancer activity of tyrosine kinase inhibitors by strengthening MAPK signaling inhibition.
[78]
AD Saleh , BA Simone , J Palazzo , JE Savage , Y Sano , T Dan , et al.
Caloric restriction augments radiation efficacy in breast cancer.
[79]
S Di Biase , HS Shim , KH Kim , M Vinciguerra , F Rappa , M Wei , et al.
Fasting regulates EGR1 and protects from glucose- and dexamethasone-dependent sensitization to chemotherapy.
[80]
KL Tinkum , KM Stemler , LS White , AJ Loza , S Jeter-Jones , BM Michalski , et al.
Fasting protects mice from lethal DNA damage by promoting small intestinal epithelial stem cell survival.
[81]
C Lee , FM Safdie , L Raffaghello , M Wei , F Madia , E Parrella , et al.
Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index.
[82]
P Sun , H Wang , Z He , X Chen , Q Wu , W Chen , et al.
Fasting inhibits colorectal cancer growth by reducing M2 polarization of tumor-associated macrophages.
[83]
F Antunes , M Corazzari , G Pereira , GM Fimia , M Piacentini , S Smaili .
Fasting boosts sensitivity of human skin melanoma to cisplatin-induced cell death.
[84]
FM Safdie , T Dorff , D Quinn , L Fontana , M Wei , C Lee , et al.
Fasting and cancer treatment in humans: A case series report.
[85]
L Raffaghello , F Safdie , G Bianchi , T Dorff , L Fontana , VD Longo .
Fasting and differential chemotherapy protection in patients.
[86]
TB Dorff , S Groshen , A Garcia , M Shah , D Tsao-Wei , H Pham , et al.
Safety and feasibility of fasting in combination with platinum-based chemotherapy.
[87]
A Michalsen , B Hoffmann , S Moebus , M Bäcker , J Langhorst , GJ Dobos .
Incorporation of fasting therapy in an integrative medicine ward: evaluation of outcome, safety, and effects on lifestyle adherence in a large prospective cohort study.
[88]
F Madeo , F Pietrocola , T Eisenberg , G Kroemer .
Caloric restriction mimetics: towards a molecular definition.
[89]
B Dwarakanath , V Jain .
Targeting glucose metabolism with 2-deoxy-D-glucose for improving cancer therapy.
[90]
VC Miranda , MI Braghiroli , LD Faria , G Bariani , A Alex , JE Bezerra Neto , et al.
Phase 2 Trial of Metformin Combined With 5-Fluorouracil in Patients With Refractory Metastatic Colorectal Cancer.
[91]
Y Liu , C He , X Huang .
Metformin partially reverses the carboplatin-resistance in NSCLC by inhibiting glucose metabolism.
[92]
F Chiarini , C Evangelisti , JA McCubrey , AM Martelli .
Current treatment strategies for inhibiting mTOR in cancer.
[93]
J Xu , D Liu , H Niu , G Zhu , Y Xu , D Ye , et al.
Resveratrol reverses Doxorubicin resistance by inhibiting epithelial-mesenchymal transition (EMT) through modulating PTEN/Akt signaling pathway in gastric cancer.
[94]
C Wang , P Ruan , Y Zhao , X Li , J Wang , X Wu , et al.
Spermidine/spermine N1-acetyltransferase regulates cell growth and metastasis via AKT/β-catenin signaling pathways in hepatocellular and colorectal carcinoma cells.
Oncotarget, 8 (2017), pp. 1092-1109
[95]
S Mou , Z Zhou , Y He , F Liu , L Gong .
Curcumin inhibits cell proliferation and promotes apoptosis of laryngeal cancer cells through Bcl-2 and PI3K/Akt, and by upregulating miR-15a.
[96]
S de Groot , MP Vreeswijk , MJ Welters , G Gravesteijn , JJ Boei , A Jochems , et al.
The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy in HER2-negative breast cancer patients: a randomized pilot study.

No potential conflict of interest was reported.

Commemorative Edition: 10 years of ICESP

Copyright © 2018. CLINICS
Article options
Tools
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos