401
|
Lipid Metabolism at the Nexus of Diet and Tumor Microenvironment. Trends Cancer 2019; 5:693-703. [PMID: 31735288 DOI: 10.1016/j.trecan.2019.09.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/27/2022]
Abstract
Obesity is a leading contributing factor to cancer development worldwide. Epidemiological evidence suggests that diet affects cancer risk and also substantially alters therapeutic outcome. Therefore, studying the impact of diet in the development and treatment of cancer should be a clinical priority. In this Review, we set out the evidence supporting the role of lipid metabolism in shaping the tumor microenvironment (TME) and cancer cell phenotype. We will discuss how dietary lipids can impact phenotype thereby affecting disease trajectory and treatment response. Finally, we will posit potential strategies on how this knowledge can be exploited to increase treatment efficacy and patient survival.
Collapse
|
402
|
|
403
|
Borsari C, Rageot D, Beaufils F, Bohnacker T, Keles E, Buslov I, Melone A, Sele AM, Hebeisen P, Fabbro D, Hillmann P, Wymann MP. Preclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl-Pyrimidine Moiety. ACS Med Chem Lett 2019; 10:1473-1479. [PMID: 31620236 PMCID: PMC6792169 DOI: 10.1021/acsmedchemlett.9b00333] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022] Open
Abstract
![]()
The
phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin
(mTOR) pathway is a critical regulator of cell growth and is frequently
hyperactivated in cancer. Therefore, PI3K inhibitors represent a valuable
asset in cancer therapy. Herein we have developed a novel anticancer
agent, the potent pan-PI3K inhibitor PQR514 (4), which
is a follow-up compound for the phase-II clinical compound PQR309
(1). Compound 4 has an improved potency
both in vitro and in cellular assays with respect to its predecessor
compounds. It shows superiority in the suppression of cancer cell
proliferation and demonstrates significant antitumor activity in an
OVCAR-3 xenograft model at concentrations approximately eight times
lower than PQR309 (1). The favorable pharmacokinetic
profile and a minimal brain penetration promote PQR514 (4) as an optimized candidate for the treatment of systemic tumors.
Collapse
Affiliation(s)
- Chiara Borsari
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Denise Rageot
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Florent Beaufils
- PIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, Switzerland
| | - Thomas Bohnacker
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Erhan Keles
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Ivan Buslov
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Anna Melone
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Alexander M. Sele
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Paul Hebeisen
- PIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, Switzerland
| | - Doriano Fabbro
- PIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, Switzerland
| | - Petra Hillmann
- PIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, Switzerland
| | - Matthias P. Wymann
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
404
|
Yan L, Raj P, Yao W, Ying H. Glucose Metabolism in Pancreatic Cancer. Cancers (Basel) 2019; 11:cancers11101460. [PMID: 31569510 PMCID: PMC6826406 DOI: 10.3390/cancers11101460] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers, with a five-year survival rate of around 5% to 8%. To date, very few available drugs have been successfully used to treat PDAC due to the poor understanding of the tumor-specific features. One of the hallmarks of pancreatic cancer cells is the deregulated cellular energetics characterized by the “Warburg effect”. It has been known for decades that cancer cells have a dramatically increased glycolytic flux even in the presence of oxygen and normal mitochondrial function. Glycolytic flux is the central carbon metabolism process in all cells, which not only produces adenosine triphosphate (ATP) but also provides biomass for anabolic processes that support cell proliferation. Expression levels of glucose transporters and rate-limiting enzymes regulate the rate of glycolytic flux. Intermediates that branch out from glycolysis are responsible for redox homeostasis, glycosylation, and biosynthesis. Beyond enhanced glycolytic flux, pancreatic cancer cells activate nutrient salvage pathways, which includes autophagy and micropinocytosis, from which the generated sugars, amino acids, and fatty acids are used to buffer the stresses induced by nutrient deprivation. Further, PDAC is characterized by extensive metabolic crosstalk between tumor cells and cells in the tumor microenvironment (TME). In this review, we will give an overview on recent progresses made in understanding glucose metabolism-related deregulations in PDAC.
Collapse
Affiliation(s)
- Liang Yan
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Priyank Raj
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wantong Yao
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
405
|
Nixon MJ, Formisano L, Mayer IA, Estrada MV, González-Ericsson PI, Isakoff SJ, Forero-Torres A, Won H, Sanders ME, Solit DB, Berger MF, Cantley LC, Winer EP, Arteaga CL, Balko JM. PIK3CA and MAP3K1 alterations imply luminal A status and are associated with clinical benefit from pan-PI3K inhibitor buparlisib and letrozole in ER+ metastatic breast cancer. NPJ Breast Cancer 2019; 5:31. [PMID: 31552290 PMCID: PMC6757060 DOI: 10.1038/s41523-019-0126-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 08/28/2019] [Indexed: 12/23/2022] Open
Abstract
Clinical trials have demonstrated the efficacy of combining phosphoinositide 3-kinase (PI3K) inhibitors with endocrine therapies in hormone therapy-refractory breast cancer. However, biomarkers of PI3K pathway dependence in ER+ breast cancer have not been fully established. Hotspot mutations in the alpha isoform of PI3K (PIK3CA) are frequent in ER+ disease and may identify tumors that respond to PI3K inhibitors. It is unclear whether PIK3CA mutations are the only biomarker to suggest pathway dependence and response to therapy. We performed correlative molecular characterization of primary and metastatic tissue from patients enrolled in a phase Ib study combining buparlisib (NVP-BKM-120), a pan-PI3K inhibitor, with letrozole in ER+, human epidermal growth factor-2 (HER2)-negative, metastatic breast cancer. Activating mutations in PIK3CA and inactivating MAP3K1 mutations marked tumors from patients with clinical benefit (≥6 months of stable disease). Patients harboring mutations in both genes exhibited the greatest likelihood of clinical benefit. In ER+ breast cancer cell lines, siRNA-mediated knockdown of MAP3K1 did not affect the response to buparlisib. In a subset of patients treated with buparlisib or the PI3Kα inhibitor alpelisib each with letrozole where PAM50 analysis was performed, nearly all tumors from patients with clinical benefit had a luminal A subtype. Mutations in MAP3K1 in ER+ breast cancer may be associated with clinical benefit from combined inhibition of PI3K and ER, but we could not ascribe direct biological function therein, suggesting they may be a surrogate for luminal A status. We posit that luminal A tumors may be a target population for this therapeutic combination.
Collapse
Affiliation(s)
- Mellissa J. Nixon
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - Luigi Formisano
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - Ingrid A. Mayer
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - M. Valeria Estrada
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - Paula I. González-Ericsson
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - Steven J. Isakoff
- Department of Medicine, Massachusetts General Hospital, Boston, MA USA
| | | | - Helen Won
- Memorial Sloan Kettering Cancer Center New York, New York, NY USA
| | - Melinda E. Sanders
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - David B. Solit
- Memorial Sloan Kettering Cancer Center New York, New York, NY USA
| | | | | | | | - Carlos L. Arteaga
- Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX USA
| | - Justin M. Balko
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
406
|
Daniel CR, McQuade JL. Nutrition and Cancer in the Microbiome Era. Trends Cancer 2019; 5:521-524. [PMID: 31474355 DOI: 10.1016/j.trecan.2019.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 02/03/2023]
Abstract
Popular views of what constitutes a healthy diet may shift, but the professional consensus is comparatively static and largely supported by our growing understanding of diet-microbiota interactions. The gut microbiome's responsiveness to diet and ability to calibrate immune and metabolic function shape new research opportunities across the cancer continuum.
Collapse
Affiliation(s)
- Carrie R Daniel
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
407
|
Castillo‐Armengol J, Fajas L, Lopez‐Mejia IC. Inter-organ communication: a gatekeeper for metabolic health. EMBO Rep 2019; 20:e47903. [PMID: 31423716 PMCID: PMC6726901 DOI: 10.15252/embr.201947903] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022] Open
Abstract
Multidirectional interactions between metabolic organs in the periphery and the central nervous system have evolved concomitantly with multicellular organisms to maintain whole-body energy homeostasis and ensure the organism's adaptation to external cues. These interactions are altered in pathological conditions such as obesity and type 2 diabetes. Bioactive peptides and proteins, such as hormones and cytokines, produced by both peripheral organs and the central nervous system, are key messengers in this inter-organ communication. Despite the early discovery of the first hormones more than 100 years ago, recent studies taking advantage of novel technologies have shed light on the multiple ways used by cells in the body to communicate and maintain energy balance. This review briefly summarizes well-established concepts and focuses on recent advances describing how specific proteins and peptides mediate the crosstalk between gut, brain, and other peripheral metabolic organs in order to maintain energy homeostasis. Additionally, this review outlines how the improved knowledge about these inter-organ networks is helping us to redefine therapeutic strategies in an effort to promote healthy living and fight metabolic disorders and other diseases.
Collapse
Affiliation(s)
| | - Lluis Fajas
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | | |
Collapse
|
408
|
Abby Philips C, Agarwal M, Phadke N, Rajesh S, Padsalgi G, Ahamed R, Augustine P. A Novel Phosphoinositide-3-kinase Adapter Protein 1 Gene Missense Mutation in Familial Cirrhosis. J Clin Exp Hepatol 2019; 9:652-656. [PMID: 31695254 PMCID: PMC6823681 DOI: 10.1016/j.jceh.2019.02.002] [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: 10/25/2018] [Accepted: 02/03/2019] [Indexed: 12/12/2022] Open
Abstract
Familial cirrhosis is a condition that is associated with the presence of liver disease with genetic linkage among multiple family members in a generation or in multiple generations. With cirrhosis, most of these disease pathogeneses are related to a defect of an enzyme/transport protein leading to a deranged metabolic pathway with variable prevalence. Many studies and high-quality metanalyses have shed light on genetic linkage associated with nonalcoholic fatty liver disease and steatohepatitis such as the PNPLA3, MBOAT7, and TM6SF2 variants. In this report, we shed light on a novel missense mutation associated with cirrhosis in a family of brothers associated with phosphoinositide-3-kinase adapter protein 1 gene through high-output whole exosome gene sequencing methodology.
Collapse
Key Words
- CCDS, Consensus Coding Sequence
- DNA, Deoxyribonucleic acid
- EWAS, Epigenome wide association study
- GWAS, Genome-wide association studies
- HCC, Hepatocellular carcinoma
- HGVS, Human Genome Variation Society
- MBOAT7, Membrane bound O-acyltransferase domain-containing 7
- NAFLD, Non-alcoholic fatty liver disease
- NGS, Next generation sequencing
- OMIM, Online Mendelian Inheritance in Man
- PBC, Primary biliary cholangitis
- PI3K
- PIK3AP1, Phosphoinositide-3-Kinase Adapter Protein 1
- PNPLA3
- PNPLA3, Patatin-like phospholipase domain containing 3
- RNA, Ribosomal nucleic acid
- RefSeq, Reference Sequence Database
- TMC4, Transmembrane channel-like 4 gene
- chronic liver disease
- epigenetics
- exosome
- familial cirrhosis
- gene mutation
- genomics
- illumina
- linkage
- metagenome
- missense mutation
Collapse
Affiliation(s)
- Cyriac Abby Philips
- The Liver Unit, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | | | - Nikhil Phadke
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, Maharashtra, India
| | - Sasidharan Rajesh
- The Liver Unit, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Guruprasad Padsalgi
- The Liver Unit, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Rizwan Ahamed
- The Liver Unit, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Philip Augustine
- The Liver Unit, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| |
Collapse
|
409
|
Genomic Evidence for Local Adaptation of Hunter-Gatherers to the African Rainforest. Curr Biol 2019; 29:2926-2935.e4. [DOI: 10.1016/j.cub.2019.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/26/2019] [Accepted: 07/04/2019] [Indexed: 12/18/2022]
|
410
|
Buchanan CM, Lee KL, Shepherd PR. For Better or Worse: The Potential for Dose Limiting the On-Target Toxicity of PI 3-Kinase Inhibitors. Biomolecules 2019; 9:biom9090402. [PMID: 31443495 PMCID: PMC6770514 DOI: 10.3390/biom9090402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/15/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
The hyper-activation of the phosphoinositide (PI) 3-kinase signaling pathway is a hallmark of many cancers and overgrowth syndromes, and as a result, there has been intense interest in the development of drugs that target the various isoforms of PI 3-kinase. Given the key role PI 3-kinases play in many normal cell functions, there is significant potential for the disruption of essential cellular functions by PI 3-kinase inhibitors in normal tissues; so-called on-target drug toxicity. It is, therefore, no surprise that progress within the clinical development of PI 3-kinase inhibitors as single-agent anti-cancer therapies has been slowed by the difficulty of identifying a therapeutic window. The aim of this review is to place the cellular, tissue and whole-body effects of PI 3-kinase inhibition in the context of understanding the potential for dose limiting on-target toxicities and to introduce possible strategies to overcome these.
Collapse
Affiliation(s)
- Christina M Buchanan
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Kate L Lee
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
411
|
|
412
|
Hsieh MH, Choe JH, Gadhvi J, Kim YJ, Arguez MA, Palmer M, Gerold H, Nowak C, Do H, Mazambani S, Knighton JK, Cha M, Goodwin J, Kang MK, Jeong JY, Lee SY, Faubert B, Xuan Z, Abel ED, Scafoglio C, Shackelford DB, Minna JD, Singh PK, Shulaev V, Bleris L, Hoyt K, Kim J, Inoue M, DeBerardinis RJ, Kim TH, Kim JW. p63 and SOX2 Dictate Glucose Reliance and Metabolic Vulnerabilities in Squamous Cell Carcinomas. Cell Rep 2019; 28:1860-1878.e9. [PMID: 31412252 PMCID: PMC7048935 DOI: 10.1016/j.celrep.2019.07.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/17/2019] [Accepted: 07/11/2019] [Indexed: 12/21/2022] Open
Abstract
Squamous cell carcinoma (SCC), a malignancy arising across multiple anatomical sites, is responsible for significant cancer mortality due to insufficient therapeutic options. Here, we identify exceptional glucose reliance among SCCs dictated by hyperactive GLUT1-mediated glucose influx. Mechanistically, squamous lineage transcription factors p63 and SOX2 transactivate the intronic enhancer cluster of SLC2A1. Elevated glucose influx fuels generation of NADPH and GSH, thereby heightening the anti-oxidative capacity in SCC tumors. Systemic glucose restriction by ketogenic diet and inhibiting renal glucose reabsorption with SGLT2 inhibitor precipitate intratumoral oxidative stress and tumor growth inhibition. Furthermore, reduction of blood glucose lowers blood insulin levels, which suppresses PI3K/AKT signaling in SCC cells. Clinically, we demonstrate a robust correlation between blood glucose concentration and worse survival among SCC patients. Collectively, this study identifies the exceptional glucose reliance of SCC and suggests its candidacy as a highly vulnerable cancer type to be targeted by systemic glucose restriction.
Collapse
Affiliation(s)
- Meng-Hsiung Hsieh
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Joshua H Choe
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jashkaran Gadhvi
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Yoon Jung Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Marcus A Arguez
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Madison Palmer
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Haleigh Gerold
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Chance Nowak
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Hung Do
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Simbarashe Mazambani
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Jordan K Knighton
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Matthew Cha
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Justin Goodwin
- Graduate School of Art and Sciences and School of Medicine, Yale University, New Haven, CT, USA
| | - Min Kyu Kang
- Department of Radiation Oncology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Brandon Faubert
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA; Center for Systems Biology, The University of Texas at Dallas, Richardson, TX, USA
| | - E Dale Abel
- Division of Endocrinology and Metabolism and the Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Claudio Scafoglio
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - David B Shackelford
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Departments of Medicine and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pankaj K Singh
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vladimir Shulaev
- Department of Biological Sciences, College of Science, Advanced Environmental Research Institute, University of North Texas, Denton, TX, USA
| | - Leonidas Bleris
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Kenneth Hoyt
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - James Kim
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tae Hoon Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Jung-Whan Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
413
|
Murugan AK. mTOR: Role in cancer, metastasis and drug resistance. Semin Cancer Biol 2019; 59:92-111. [PMID: 31408724 DOI: 10.1016/j.semcancer.2019.07.003] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/14/2019] [Accepted: 07/03/2019] [Indexed: 02/09/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that gets inputs from the amino acids, nutrients, growth factor, and environmental cues to regulate varieties of fundamental cellular processes which include protein synthesis, growth, metabolism, aging, regeneration, autophagy, etc. The mTOR is frequently deregulated in human cancer and activating somatic mutations of mTOR were recently identified in several types of human cancer and hence mTOR is therapeutically targeted. mTOR inhibitors were commonly used as immunosuppressors and currently, it is approved for the treatment of human malignancies. This review briefly focuses on the structure and biological functions of mTOR. It extensively discusses the genetic deregulation of mTOR including amplifications and somatic mutations, mTOR-mediated cell growth promoting signaling, therapeutic targeting of mTOR and the mechanisms of resistance, the role of mTOR in precision medicine and other recent advances in further understanding the role of mTOR in cancer.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, PO Box 3354, Research Center (MBC 03), Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
414
|
Untargeted Metabolomics Reveals Molecular Effects of Ketogenic Diet on Healthy and Tumor Xenograft Mouse Models. Int J Mol Sci 2019; 20:ijms20163873. [PMID: 31398922 PMCID: PMC6719192 DOI: 10.3390/ijms20163873] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023] Open
Abstract
The application of ketogenic diet (KD) (high fat/low carbohydrate/adequate protein) as an auxiliary cancer therapy is a field of growing attention. KD provides sufficient energy supply for healthy cells, while possibly impairing energy production in highly glycolytic tumor cells. Moreover, KD regulates insulin and tumor related growth factors (like insulin growth factor-1, IGF-1). In order to provide molecular evidence for the proposed additional inhibition of tumor growth when combining chemotherapy with KD, we applied untargeted quantitative metabolome analysis on a spontaneous breast cancer xenograft mouse model, using MDA-MB-468 cells. Healthy mice and mice bearing breast cancer xenografts and receiving cyclophosphamide chemotherapy were compared after treatment with control diet and KD. Metabolomic profiling was performed on plasma samples, applying high-performance liquid chromatography coupled to tandem mass spectrometry. Statistical analysis revealed metabolic fingerprints comprising numerous significantly regulated features in the group of mice bearing breast cancer. This fingerprint disappeared after treatment with KD, resulting in recovery to the metabolic status observed in healthy mice receiving control diet. Moreover, amino acid metabolism as well as fatty acid transport were found to be affected by both the tumor and the applied KD. Our results provide clear evidence of a significant molecular effect of adjuvant KD in the context of tumor growth inhibition and suggest additional mechanisms of tumor suppression beyond the proposed constrain in energy supply of tumor cells.
Collapse
|
415
|
Pedini F, De Luca G, Felicetti F, Puglisi R, Boe A, Arasi MB, Fratini F, Mattia G, Spada M, Caporali S, Biffoni M, Giuliani A, Carè A, Felli N. Joint action of miR-126 and MAPK/PI3K inhibitors against metastatic melanoma. Mol Oncol 2019; 13:1836-1854. [PMID: 31115969 PMCID: PMC6717748 DOI: 10.1002/1878-0261.12506] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/17/2019] [Accepted: 05/20/2019] [Indexed: 02/03/2023] Open
Abstract
Emerging data support the rationale of combined therapies in advanced melanoma. Specifically, the combined use of drugs with different mechanisms of action can reduce the probability of selecting resistant clones. To identify agents active against melanoma cells, we screened a library of 349 anti‐cancer compounds, currently in clinical use or trials, and selected PIK‐75, an inhibitor of the phosphatidylinositol 3‐kinase/protein kinase B (PI3K/AKT) pathway, as the ‘top active’ drug. PIK‐75 was then used alone or in combination with vemurafenib, the first BRAF inhibitor approved for patients with melanoma harboring BRAF mutations. We identified a combined dose of PIK‐75 and vemurafenib that inhibited both the PI3K/AKT and mitogen‐activated protein kinase pathways, thereby overcoming any compensatory activation. In view of the important tumor suppressor function induced by restoring expression of microRNA (miR)‐126 in metastatic melanoma cells, we examined whether miR‐126 has a synergistic role when included in a triple combination alongside PIK‐75 and vemurafenib. We found that enforced expression of miR‐126 (which alone can reduce tumorigenicity) significantly increased PIK‐75 activity when used as either a single agent or in combination with vemurafenib. Interestingly, PIK‐75 proved to be effective against early passage cell lines derived from patients’ biopsies and on melanoma cell lines resistant to either vemurafenib or dabrafenib, thus suggesting that it potentially has the capability to overcome drug resistance. Finally, the synergistic role played by miR‐126 in combination with vemurafenib and/or PIK‐75 was demonstrated in vivo in mouse xenograft models, in which tumor growth inhibition was associated with increased apoptosis. These results not only show the efficacy of PIK‐75 and vemurafenib co‐treatment but also indicate that restoration of miR‐126 expression in advanced melanoma can enhance their antitumor activity, which may possibly allow dose reduction to decrease adverse events without reducing the therapeutic benefits.
Collapse
Affiliation(s)
- Francesca Pedini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriele De Luca
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Federica Felicetti
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Rossella Puglisi
- Center for Gender Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Boe
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Beatrice Arasi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Gianfranco Mattia
- Center for Gender Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Caporali
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata-IRCCS, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandro Giuliani
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Carè
- Center for Gender Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Nadia Felli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
416
|
Montrose DC, Galluzzi L. Drugging cancer metabolism: Expectations vs. reality. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 347:1-26. [PMID: 31451211 DOI: 10.1016/bs.ircmb.2019.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As compared to their normal counterparts, neoplastic cells exhibit a variety of metabolic changes that reflect not only genetic and epigenetic defects underlying malignant transformation, but also the nutritional and immunobiological conditions of the tumor microenvironment. Such alterations, including the so-called Warburg effect (an increase in glucose uptake largely feeding anabolic and antioxidant metabolism), have attracted considerable attention as potential targets for the development of novel anticancer therapeutics. However, very few drugs specifically conceived to target bioenergetic cancer metabolism are currently approved by regulatory agencies for use in humans. This reflects the elevated degree of heterogeneity and redundancy in the metabolic circuitries exploited by neoplastic cells from different tumors (even of the same type), as well as the resemblance of such metabolic pathways to those employed by highly proliferating normal cells. Here, we summarize the major metabolic alterations that accompany oncogenesis, the potential of targeting bioenergetic metabolism for cancer therapy, and the obstacles that still prevent the clinical translation of such a promising therapeutic paradigm.
Collapse
Affiliation(s)
- David C Montrose
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université Paris Descartes/Paris V, Paris, France.
| |
Collapse
|
417
|
Vara-Ciruelos D, Russell FM, Hardie DG. The strange case of AMPK and cancer: Dr Jekyll or Mr Hyde? †. Open Biol 2019; 9:190099. [PMID: 31288625 PMCID: PMC6685927 DOI: 10.1098/rsob.190099] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
The AMP-activated protein kinase (AMPK) acts as a cellular energy sensor. Once switched on by increases in cellular AMP : ATP ratios, it acts to restore energy homeostasis by switching on catabolic pathways while switching off cell growth and proliferation. The canonical AMP-dependent mechanism of activation requires the upstream kinase LKB1, which was identified genetically to be a tumour suppressor. AMPK can also be switched on by increases in intracellular Ca2+, by glucose starvation and by DNA damage via non-canonical, AMP-independent pathways. Genetic studies of the role of AMPK in mouse cancer suggest that, before disease arises, AMPK acts as a tumour suppressor that protects against cancer, with this protection being further enhanced by AMPK activators such as the biguanide phenformin. However, once cancer has occurred, AMPK switches to being a tumour promoter instead, enhancing cancer cell survival by protecting against metabolic, oxidative and genotoxic stresses. Studies of genetic changes in human cancer also suggest diverging roles for genes encoding subunit isoforms, with some being frequently amplified, while others are mutated.
Collapse
Affiliation(s)
| | | | - D. Grahame Hardie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
418
|
Zhao F, Siu JJ, Huang W, Askwith C, Cao L. Insulin Modulates Excitatory Synaptic Transmission and Synaptic Plasticity in the Mouse Hippocampus. Neuroscience 2019; 411:237-254. [PMID: 31146008 PMCID: PMC6612444 DOI: 10.1016/j.neuroscience.2019.05.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
The administration of exogenous insulin into the hippocampus has the potential to enhance cognitive function and exert other beneficial effects. Elucidating the neurobiological substrates of insulin action and its underlying physiological mechanisms may further improve treatment efficacy. Previous work has shown that insulin affects synaptic plasticity, however there are discrepancies and contradictory conclusions between studies. Here, we used extracellular field recordings in mouse hippocampal slices to investigate how insulin acutely modulates synaptic transmission and synaptic plasticity, both of which are correlated with learning and memory processes. Our data demonstrate that insulin application inhibited basal excitatory synaptic transmission and promoted long-term potentiation (LTP) induction at hippocampal Schaffer collateral-CA1 synapses. Under similar conditions, insulin strongly activated the PI3K/AKT pathway, but had only a weak effect on the MAPK/ERK pathway. Although insulin-induced inhibition of field excitatory post-synaptic potentials (fEPSPs) was previously termed insulin-long-term depression (insulin-LTD), insulin application potentiated recovery from classically induced LTD. Further analysis suggests suppression of presynaptic neurotransmitter release contributed to the insulin-LTD. At low concentrations, insulin primarily inhibited fEPSPs; however, at high concentration, its effects were of mixed inhibition and enhancement in different recordings. Moreover, a broad spectrum protein kinase C blocker, cannabinoid receptor type 1 activator, or a high glucose concentration inhibited fEPSPs per se, and disturbed insulin's effect on fEPSP. Insulin also caused depotentiation during LTP expression and triggered depression during LTD recovery. Given the essential roles of dynamic synaptic transmission and plasticity in learning and memory, our data provide more evidence that insulin application may actively modulate hippocampal-dependent cognitive events.
Collapse
Affiliation(s)
- Fangli Zhao
- College of Medicine, The Ohio State University
| | - Jason J Siu
- College of Medicine, The Ohio State University
| | - Wei Huang
- College of Medicine, The Ohio State University
| | | | - Lei Cao
- College of Medicine, The Ohio State University.
| |
Collapse
|
419
|
Abstract
Most cancer patients die due to metastasis formation. Therefore, understanding, preventing, and treating metastatic cancers is an unmet need. Recent research indicates that cancer cells that undergo metastasis formation have a distinct metabolism that can be targeted. Here, I would like to discuss potential opportunities in exploiting the metabolic vulnerabilities of metastasizing cancer cells.
Collapse
Affiliation(s)
- Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000, Leuven, Belgium. .,Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
420
|
De Angelis ML, Francescangeli F, La Torre F, Zeuner A. Stem Cell Plasticity and Dormancy in the Development of Cancer Therapy Resistance. Front Oncol 2019; 9:626. [PMID: 31355143 PMCID: PMC6636659 DOI: 10.3389/fonc.2019.00626] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer treatment with either standard chemotherapy or targeted agents often results in the emergence of drug-refractory cell populations, ultimately leading to therapy failure. The biological features of drug resistant cells are largely overlapping with those of cancer stem cells and include heterogeneity, plasticity, self-renewal ability, and tumor-initiating capacity. Moreover, drug resistance is usually characterized by a suppression of proliferation that can manifest as quiescence, dormancy, senescence, or proliferative slowdown. Alterations in key cellular pathways such as autophagy, unfolded protein response or redox signaling, as well as metabolic adaptations also contribute to the establishment of drug resistance, thus representing attractive therapeutic targets. Moreover, a complex interplay of drug resistant cells with the micro/macroenvironment and with the immune system plays a key role in dictating and maintaining the resistant phenotype. Recent studies have challenged traditional views of cancer drug resistance providing innovative perspectives, establishing new connections between drug resistant cells and their environment and indicating unexpected therapeutic strategies. In this review we discuss recent advancements in understanding the mechanisms underlying drug resistance and we report novel targeting agents able to overcome the drug resistant status, with particular focus on strategies directed against dormant cells. Research on drug resistant cancer cells will take us one step forward toward the development of novel treatment approaches and the improvement of relapse-free survival in solid and hematological cancer patients.
Collapse
Affiliation(s)
- Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Filippo La Torre
- Department of Surgical Sciences Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
421
|
Abstract
PURPOSE OF REVIEW The ketogenic diet, a high-fat, low-carbohydrate therapy, has become an established treatment for pediatric epilepsy since 1921. There has recently been an increase in important studies on the ketogenic diet, and this review will highlight the most recent in order to provide a synthesis of where this field stands today. RECENT FINDINGS Clinical studies continue to support the use of ketogenic diets in epilepsy, with more recent trials supporting its use in adults. Clinical recommendations published in 2018 based on a decade of practice and research, guide implementation and management of the ketogenic diet in epilepsy. One of the most rapidly growing 'indications' includes the role of ketogenic diets in status epilepticus. An exciting new potential mechanism for how the ketogenic diet exerts its antiseizure effects is through changing the composition of the gut microbiome. Lastly, ketogenic diets are being applied to a range of neurological conditions from autism to Alzheimer's disease. SUMMARY The ketogenic diet is a versatile therapy, with growing clinical evidence and guidelines, widely used for the treatment of epilepsy. New indications include status epilepticus and neurological conditions other than epilepsy.
Collapse
Affiliation(s)
- Danielle M deCampo
- Departments of Neurology and Pediatrics, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | |
Collapse
|
422
|
Modifiable Host Factors in Melanoma: Emerging Evidence for Obesity, Diet, Exercise, and the Microbiome. Curr Oncol Rep 2019; 21:72. [PMID: 31263961 DOI: 10.1007/s11912-019-0814-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW We discuss how potentially modifiable factors including obesity, the microbiome, diet, and exercise may impact melanoma development, progression, and therapeutic response. RECENT FINDINGS Obesity is unexpectedly associated with improved outcomes with immune and targeted therapy in melanoma, with early mechanistic data suggesting leptin as one mediator. The gut microbiome is both a biomarker of response to immunotherapy and a potential target. As diet is a major determinant of the gut microbiome, ongoing studies are examining the interaction between diet, the gut microbiome, and immunity. Data are emerging for a potential role of exercise in reducing hypoxia and enhancing anti-tumor immunity, though this has not yet been well-studied in the context of contemporary therapies. Recent data suggests energy balance may play a role in the outcomes of metastatic melanoma. Further studies are needed to demonstrate mechanism and causality as well as the feasibility of targeting these factors.
Collapse
|
423
|
Rageot D, Bohnacker T, Keles E, McPhail JA, Hoffmann RM, Melone A, Borsari C, Sriramaratnam R, Sele AM, Beaufils F, Hebeisen P, Fabbro D, Hillmann P, Burke JE, Wymann MP. ( S)-4-(Difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530), a Potent, Orally Bioavailable, and Brain-Penetrable Dual Inhibitor of Class I PI3K and mTOR Kinase. J Med Chem 2019; 62:6241-6261. [PMID: 31244112 DOI: 10.1021/acs.jmedchem.9b00525] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is frequently overactivated in cancer, and drives cell growth, proliferation, survival, and metastasis. Here, we report a structure-activity relationship study, which led to the discovery of a drug-like adenosine 5'-triphosphate-site PI3K/mTOR kinase inhibitor: (S)-4-(difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530, compound 6), which qualifies as a clinical candidate due to its potency and specificity for PI3K and mTOR kinases, and its pharmacokinetic properties, including brain penetration. Compound 6 showed excellent selectivity over a wide panel of kinases and an excellent selectivity against unrelated receptor enzymes and ion channels. Moreover, compound 6 prevented cell growth in a cancer cell line panel. The preclinical in vivo characterization of compound 6 in an OVCAR-3 xenograft model demonstrated good oral bioavailability, excellent brain penetration, and efficacy. Initial toxicity studies in rats and dogs qualify 6 for further development as a therapeutic agent in oncology.
Collapse
Affiliation(s)
- Denise Rageot
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Thomas Bohnacker
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Erhan Keles
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Jacob A McPhail
- Department of Biochemistry and Microbiology , University of Victoria , Victoria , British Columbia V8W 2Y2 , Canada
| | - Reece M Hoffmann
- Department of Biochemistry and Microbiology , University of Victoria , Victoria , British Columbia V8W 2Y2 , Canada
| | - Anna Melone
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Chiara Borsari
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Rohitha Sriramaratnam
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Alexander M Sele
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Florent Beaufils
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| | - Paul Hebeisen
- PIQUR Therapeutics AG , Hochbergerstrasse 60C , 4057 Basel , Switzerland
| | - Doriano Fabbro
- PIQUR Therapeutics AG , Hochbergerstrasse 60C , 4057 Basel , Switzerland
| | - Petra Hillmann
- PIQUR Therapeutics AG , Hochbergerstrasse 60C , 4057 Basel , Switzerland
| | - John E Burke
- Department of Biochemistry and Microbiology , University of Victoria , Victoria , British Columbia V8W 2Y2 , Canada
| | - Matthias P Wymann
- Department of Biomedicine , University of Basel , Mattenstrasse 28 , 4058 Basel , Switzerland
| |
Collapse
|
424
|
Zhang Y, Earp HS, Liu P. Beyond growth signaling: apoptotic sensor MERTK activates AKT by a novel mechanism. Mol Cell Oncol 2019; 6:1611161. [PMID: 31211243 DOI: 10.1080/23723556.2019.1611161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 01/11/2023]
Abstract
Canonically the oncogenic kinase AKT is activated by growth signals. Our work suggests apoptotic materials, abundant in tumors, also contribute to AKT activation by stimulating MERTK that in turn phosphorylates Y26 in the AKT PH domain. pY26 reverses binding of an AKT endogenous, WW-domain containing inhibitor, SAV1, allowing AKT responsiveness to classic growth signals. This novel mechanism may contribute to drug resistance.
Collapse
Affiliation(s)
- Yanqiong Zhang
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - H Shelton Earp
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Medicine and Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pengda Liu
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
425
|
Molinaro A, Becattini B, Mazzoli A, Bleve A, Radici L, Maxvall I, Sopasakis VR, Molinaro A, Bäckhed F, Solinas G. Insulin-Driven PI3K-AKT Signaling in the Hepatocyte Is Mediated by Redundant PI3Kα and PI3Kβ Activities and Is Promoted by RAS. Cell Metab 2019; 29:1400-1409.e5. [PMID: 30982732 DOI: 10.1016/j.cmet.2019.03.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 02/04/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Phosphatidylinositol-3-kinase (PI3K) activity is aberrant in tumors, and PI3K inhibitors are investigated as cancer therapeutics. PI3K signaling mediates insulin action in metabolism, but the role of PI3K isoforms in insulin signaling remains unresolved. Defining the role of PI3K isoforms in insulin signaling is necessary for a mechanistic understanding of insulin action and to develop PI3K inhibitors with optimal therapeutic index. We show that insulin-driven PI3K-AKT signaling depends on redundant PI3Kα and PI3Kβ activities, whereas PI3Kδ and PI3Kγ are largely dispensable. We have also found that RAS activity promotes AKT phosphorylation in insulin-stimulated hepatocytes and that promotion of insulin-driven AKT phosphorylation by RAS depends on PI3Kα. These findings reveal the detailed mechanism by which insulin activates AKT, providing an improved mechanistic understanding of insulin signaling. This improved model for insulin signaling predicts that isoform-selective PI3K inhibitors discriminating between PI3Kα and PI3Kβ should be dosed below their hyperglycemic threshold to achieve isoform selectivity.
Collapse
Affiliation(s)
- Angela Molinaro
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Barbara Becattini
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Arianna Mazzoli
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Augusto Bleve
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Lucia Radici
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ingela Maxvall
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Victoria Rotter Sopasakis
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Antonio Molinaro
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Giovanni Solinas
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
426
|
Bland JS. What is Evidence-Based Functional Medicine in the 21st Century? Integr Med (Encinitas) 2019; 18:14-18. [PMID: 32549804 PMCID: PMC7217393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The 21st century has already demonstrated itself to be an era of change for medicine and science. There is a new openness-to ideas, to a shift in perspectives, to a redefinition of evidence and the many ways it can be gathered. New interest in real-world data, patient-experience information has also become an increasingly important contributor to the evaluation of treatment effectiveness. It is a fertile time on many fronts, including an expanded reach for a systems biology formalism and the Functional Medicine movement.
Collapse
|
427
|
Daurio NA, Wang Y, Chen Y, Zhou H, Carballo-Jane E, Mane J, Rodriguez CG, Zafian P, Houghton A, Addona G, McLaren DG, Zhang R, Shyong BJ, Bateman K, Downes DP, Webb M, Kelley DE, Previs SF. Spatial and temporal studies of metabolic activity: contrasting biochemical kinetics in tissues and pathways during fasted and fed states. Am J Physiol Endocrinol Metab 2019; 316:E1105-E1117. [PMID: 30912961 DOI: 10.1152/ajpendo.00459.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The regulation of nutrient homeostasis, i.e., the ability to transition between fasted and fed states, is fundamental in maintaining health. Since food is typically consumed over limited (anabolic) periods, dietary components must be processed and stored to counterbalance the catabolic stress that occurs between meals. Herein, we contrast tissue- and pathway-specific metabolic activity in fasted and fed states. We demonstrate that knowledge of biochemical kinetics that is obtained from opposite ends of the energetic spectrum can allow mechanism-based differentiation of healthy and disease phenotypes. Rat models of type 1 and type 2 diabetes serve as case studies for probing spatial and temporal patterns of metabolic activity via [2H]water labeling. Experimental designs that capture integrative whole body metabolism, including meal-induced substrate partitioning, can support an array of research surrounding metabolic disease; the relative simplicity of the approach that is discussed here should enable routine applications in preclinical models.
Collapse
Affiliation(s)
- Natalie A Daurio
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Yichen Wang
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Ying Chen
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Haihong Zhou
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Ester Carballo-Jane
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Joel Mane
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Carlos G Rodriguez
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Peter Zafian
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Andrea Houghton
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - George Addona
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - David G McLaren
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Rena Zhang
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Bao Jen Shyong
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Kevin Bateman
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Daniel P Downes
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Maria Webb
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - David E Kelley
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Stephen F Previs
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| |
Collapse
|
428
|
Sullivan MR, Vander Heiden MG. Determinants of nutrient limitation in cancer. Crit Rev Biochem Mol Biol 2019; 54:193-207. [PMID: 31162937 PMCID: PMC6715536 DOI: 10.1080/10409238.2019.1611733] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022]
Abstract
Proliferation requires that cells accumulate sufficient biomass to grow and divide. Cancer cells within tumors must acquire a variety of nutrients, and tumor growth slows or stops if necessary metabolites are not obtained in sufficient quantities. Importantly, the metabolic demands of cancer cells can be different from those of untransformed cells, and nutrient accessibility in tumors is different than in many normal tissues. Thus, cancer cell survival and proliferation may be limited by different metabolic factors than those that are necessary to maintain noncancerous cells. Understanding the variables that dictate which nutrients are critical to sustain tumor growth may identify vulnerabilities that could be used to treat cancer. This review examines the various cell-autonomous, local, and systemic factors that determine which nutrients are limiting for tumor growth.
Collapse
Affiliation(s)
- Mark R Sullivan
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology , Cambridge , MA , USA
- Dana-Farber Cancer Institute , Boston , MA , USA
| |
Collapse
|
429
|
Kaiser A, Haskins C, Siddiqui MM, Hussain A, D’Adamo C. The evolving role of diet in prostate cancer risk and progression. Curr Opin Oncol 2019; 31:222-229. [PMID: 30893147 PMCID: PMC7379157 DOI: 10.1097/cco.0000000000000519] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This overview examines the rationale for dietary interventions for prostate cancer by summarizing the current evidence base and biological mechanisms for the involvement of diet in disease incidence and progression. RECENT FINDINGS Recent data have further solidified the association between insulin resistance and prostate cancer with the homeostatic model assessment of insulin resistance. Data also show that periprostatic adipocytes promote extracapsular extension of prostate cancer through chemokines, thereby providing a mechanistic explanation for the association observed between obesity and high-grade cancer. Regarding therapeutics, hyperinsulinemia may be the cause of resistance to phosphatidylinositol-3 kinase inhibitors in the treatment of prostate cancer, leading to new investigations combining these drugs with ketogenic diets. SUMMARY Given the recently available data regarding insulin resistance and adipokine influence on prostate cancer, dietary strategies targeting metabolic syndrome, diabetes, and obesity should be further explored. In macronutrient-focused therapies, low carbohydrate/ketogenic diets should be favored in such interventions because of their superior impact on weight loss and metabolic parameters and encouraging clinical data. Micronutrients, including the carotenoid lycopene which is found in highest concentrations in tomatoes, may also play a role in prostate cancer prevention and prognosis through complementary metabolic mechanisms. The interplay between genetics, diet, and prostate cancer is an area of emerging focus that might help optimize therapeutic dietary response in the future through personalization.
Collapse
Affiliation(s)
- Adeel Kaiser
- Department of Radiation Oncology, Univ. of Maryland School of Medicine, Baltimore, MD USA
| | - Christopher Haskins
- Department of Radiation Oncology, Univ. of Maryland School of Medicine, Baltimore, MD USA
| | - Mohummad M. Siddiqui
- Division of of Urology, Department of Surgery, Univ. of Maryland School of Medicine, Baltimore, MD USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD USA
| | - Arif Hussain
- Department of Medicine and University of Maryland Greenebaum Comprehensive Cancer Center, Univ. of Maryland School of Medicine, Baltimore, MD USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD USA
| | - Christopher D’Adamo
- Department of Family and Community Medicine, Univ. of Maryland School of Medicine, Baltimore, MD USA
| |
Collapse
|
430
|
Lévesque S, Pol JG, Ferrere G, Galluzzi L, Zitvogel L, Kroemer G. Trial watch: dietary interventions for cancer therapy. Oncoimmunology 2019; 8:1591878. [PMID: 31143510 PMCID: PMC6527263 DOI: 10.1080/2162402x.2019.1591878] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/30/2022] Open
Abstract
Dietary interventions have a profound impact on whole body metabolism, including oncometabolism (the metabolic features allowing cancer cells to proliferate) and immunometabolism (the catabolic and anabolic reactions that regulate immune responses). Recent preclinical studies demonstrated that multiple dietary changes can improve anticancer immunosurveillance of chemo-, radio- and immunotherapy. These findings have fostered the design of clinical trials evaluating the capacity of dietary interventions to synergize with treatment and hence limit tumor progression. Here, we discuss the scientific rationale for harnessing dietary interventions to improve the efficacy of anticancer therapy and present up-to-date information on clinical trials currently investigating this possibility.
Collapse
Affiliation(s)
- Sarah Lévesque
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Orsay, France.,Fondation pour la Recherche Médicale, Paris, France
| | - Jonathan G Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Gladys Ferrere
- INSERM U1015, Villejuif, France.,CICBT507, Villejuif, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Laurence Zitvogel
- Université Paris-Saclay, Orsay, France.,INSERM U1015, Villejuif, France.,CICBT507, Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Orsay, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
431
|
Rizzieri D, Paul B, Kang Y. Metabolic alterations and the potential for targeting metabolic pathways in the treatment of multiple myeloma. ACTA ACUST UNITED AC 2019; 5. [PMID: 31020046 PMCID: PMC6476731 DOI: 10.20517/2394-4722.2019.05] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Metabolism is defined as the collection of complex biochemical processes that living cells use to generate energy and maintain their growth and survival. Metabolism encompasses the synthesis and breakdown of glucose, fatty acids, and amino acids; the generation of energy (ATP); and oxidative phosphorylation. In cancer cells, metabolism can be commandeered to promote tumor growth and cellular proliferation. These alterations in metabolism have emerged as an additional hallmark of various cancers. In this review we focus on metabolic alterations in multiple myeloma (MM) - a malignancy of plasma cells - including derangements in glycolysis, gluconeogenesis, the tricarboxylic acid cycle, oxidative phosphorylation, and fatty acid/amino acid synthesis and degradation. Particular focus is given to metabolic alterations that contribute to myeloma cell growth, proliferation and drug resistance. Finally, novel approaches that target metabolic pathways for the treatment of MM are discussed.
Collapse
Affiliation(s)
- Dustin Rizzieri
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Barry Paul
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Yubin Kang
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
432
|
Gillies RJ, Pilot C, Marunaka Y, Fais S. Targeting acidity in cancer and diabetes. Biochim Biophys Acta Rev Cancer 2019; 1871:273-280. [PMID: 30708040 PMCID: PMC6525044 DOI: 10.1016/j.bbcan.2019.01.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 02/08/2023]
Abstract
While cancer is commonly described as "a disease of the genes", it is also a disease of metabolism. Indeed, carcinogenesis and malignancy are highly associated with metabolic re-programming, and there is clinical evidence that interrupting a cancer's metabolic program can improve patients' outcomes. Notably, many of the metabolic adaptations observed in cancer are similar to the same perturbations observed in diabetic patients. For example, metformin is commonly used to reduce hyperglycemia in diabetic patients, and has been demonstrated to reduce cancer incidence. Treatment with PI3K inhibitors can induce hyperinsulinemia, which can blunt therapeutic efficacy if unchecked. While commonalities between metabolism in cancer and diabetes have been extensively reviewed, here we examine a less explored and emergent convergence between diabetic and cancer metabolism: the generation of lactic acid and subsequent acidification of the surrounding microenvironment. Extracellular lactic acidosis is integral in disease manifestation and is a negative prognostic in both disease states. In tumors, this results in important sequela for cancer progression including increased invasion and metastasis, as well as inhibition of immune surveillance. In diabetes, acidosis impacts the ability of insulin to bind to its receptor, leading to peripheral resistance and an exacerbation of symptoms. Thus, acidosis may be a relevant therapeutic target, and we describe three approaches for targeting: buffers, nanomedicine, and proton pump inhibitors.
Collapse
Affiliation(s)
- Robert J Gillies
- Dept. Cancer Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33602, USA.
| | - Christian Pilot
- Dept. Cancer Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33602, USA
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto 604-8472, Japan; Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu 525-8577, Japan.
| | - Stefano Fais
- Dept. of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Roma 00161, Italy.
| |
Collapse
|
433
|
De Santis MC, Gulluni F, Campa CC, Martini M, Hirsch E. Targeting PI3K signaling in cancer: Challenges and advances. Biochim Biophys Acta Rev Cancer 2019; 1871:361-366. [DOI: 10.1016/j.bbcan.2019.03.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022]
|
434
|
Paddock MN, Field SJ, Cantley LC. Treating cancer with phosphatidylinositol-3-kinase inhibitors: increasing efficacy and overcoming resistance. J Lipid Res 2019; 60:747-752. [PMID: 30718284 PMCID: PMC6446698 DOI: 10.1194/jlr.s092130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/25/2019] [Indexed: 11/20/2022] Open
Abstract
The discovery of the phosphatidylinositol-3-kinase (PI3K) pathway was a major advance in understanding growth factor signaling. The high frequency of PI3K pathway mutations in many cancers has encouraged a new field targeting cancer driver mutations. Although there have been many successes, targeting PI3K itself has proven challenging, in part because of its multiple isoforms with distinct roles. Despite promising preclinical results, development of PI3K inhibitors as pharmacologic anticancer agents has been limited by modest single-agent efficacy and significant adverse effects. If we could overcome these limitations, PI3K inhibitors would be a powerful cancer-fighting tool. Data from phase III clinical trials yields insight into some of the problems with PI3K inhibitors. Recent advances have shed light on the mechanisms of tumor resistance to PI3K inhibitors via feedback pathways that cause elevated insulin levels that then activate the same PI3K pathways that are the targets of inhibition. Improving our understanding of the complex regulatory feedback pathways that activate in response to PI3K inhibition will reveal ways to increase the efficacy of PI3K inhibitors and reduce adverse effects, increasing the usefulness of this class as a treatment option for multiple cancer types.
Collapse
Affiliation(s)
- Marcia N Paddock
- Meyer Cancer Center Weill Cornell Medicine, New York, NY 10021; Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY 10021
| | - Seth J Field
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093.
| | - Lewis C Cantley
- Meyer Cancer Center Weill Cornell Medicine, New York, NY 10021.
| |
Collapse
|
435
|
Hanker AB, Kaklamani V, Arteaga CL. Challenges for the Clinical Development of PI3K Inhibitors: Strategies to Improve Their Impact in Solid Tumors. Cancer Discov 2019; 9:482-491. [PMID: 30867161 PMCID: PMC6445714 DOI: 10.1158/2159-8290.cd-18-1175] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/04/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
The PI3K pathway is mutated and aberrantly activated in many cancers and plays a central role in tumor cell proliferation and survival, making it a rational therapeutic target. Until recently, however, results from clinical trials with PI3K inhibitors in solid tumors have been largely disappointing. Here, we describe several factors that have limited the success of these agents, including the weak driver oncogenic activity of mutant PI3K, suboptimal patient selection in trials, drug-related toxicities, feedback upregulation of compensatory mechanisms when PI3K is blocked, increased insulin production upon PI3Kα inhibition, lack of mutant-specific inhibitors, and a relative scarcity of studies using combinations with PI3K antagonists. We also suggest strategies to improve the impact of these agents in solid tumors. Despite these challenges, we are optimistic that isoform-specific PI3K inhibitors, particularly in combination with other agents, may be valuable in treating appropriately selected patients with PI3K-dependent tumors. SIGNIFICANCE: Despite the modest clinical activity of PI3K inhibitors in solid tumors, there is an increasing understanding of the factors that may have limited their success. Strategies to ameliorate drug-related toxicities, use of rational combinations with PI3K antagonists, development of mutant-selective PI3K inhibitors, and better patient selection should improve the success of these targeted agents against solid tumors.
Collapse
Affiliation(s)
- Ariella B. Hanker
- Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | | | - Carlos L. Arteaga
- Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
436
|
Cuyàs E, Buxó M, Ferri Iglesias MJ, Verdura S, Pernas S, Dorca J, Álvarez I, Martínez S, Pérez-Garcia JM, Batista-López N, Rodríguez-Sánchez CA, Amillano K, Domínguez S, Luque M, Morilla I, Stradella A, Viñas G, Cortés J, Joven J, Brunet J, López-Bonet E, Garcia M, Saidani S, Queralt Moles X, Martin-Castillo B, Menendez JA. The C Allele of ATM rs11212617 Associates With Higher Pathological Complete Remission Rate in Breast Cancer Patients Treated With Neoadjuvant Metformin. Front Oncol 2019; 9:193. [PMID: 30984619 PMCID: PMC6447648 DOI: 10.3389/fonc.2019.00193] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/06/2019] [Indexed: 12/21/2022] Open
Abstract
Background: The minor allele (C) of the single-nucleotide polymorphism (SNP) rs11212617, located near the ataxia telangiectasia mutated (ATM) gene, has been associated with an increased likelihood of treatment success with metformin in type 2 diabetes. We herein investigated whether the same SNP would predict clinical response to neoadjuvant metformin in women with early breast cancer (BC). Methods: DNA was collected from 79 patients included in the intention-to-treat population of the METTEN study, a phase 2 clinical trial of HER2-positive BC patients randomized to receive either metformin combined with anthracycline/taxane-based chemotherapy and trastuzumab or equivalent regimen without metformin, before surgery. SNP rs11212617 genotyping was assessed using allelic discrimination by quantitative polymerase chain reaction. Results: Logistic regression analyses revealed a significant relationship between the rs11212617 genotype and the ability of treatment arms to achieve a pathological complete response (pCR) in patients (odds ratio [OR]genotype×arm = 10.33, 95% confidence interval [CI]: 1.29-82.89, p = 0.028). In the metformin-containing arm, patients bearing the rs11212617 C allele had a significantly higher probability of pCR (OR A/C,C/C = 7.94, 95%CI: 1.60-39.42, p = 0.011). Conversely, no association was found between rs11212617 and clinical response in the reference arm (OR A/C,C/C = 0.77, 95%CI: 0.20-2.92, p = 0.700). After controlling for tumor size and hormone receptor status, the rs11212617 C allele remained a significant predictor of pCR solely in the metformin-containing arm. Conclusions: If reproducible, the rs11212617 C allele might warrant consideration as a predictive clinical biomarker to inform the personalized use of metformin in BC patients. Trial Registration: EU Clinical Trials Register, EudraCT number 2011-000490-30. Registered 28 February 2011, https://www.clinicaltrialsregister.eu/ctr-search/trial/2011-000490-30/ES.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Maria Buxó
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | | | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Sonia Pernas
- Breast Unit, Department of Medical Oncology, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Joan Dorca
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain
| | - Isabel Álvarez
- Medical Oncology Service, Hospital Universitario Donostia, Donostia-San Sebastián, Spain.,Biodonostia Health Research Institute, Donostia-San Sebastián, Spain
| | - Susana Martínez
- Medical Oncology Department, Hospital de Mataró, Mataró, Barcelona, Spain
| | | | - Norberto Batista-López
- Medical Oncology Service, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - César A Rodríguez-Sánchez
- Medical Oncology Service, Hospital Universitario de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Kepa Amillano
- Medical Oncology, Hospital Universitari Sant Joan, Reus, Spain
| | - Severina Domínguez
- Medical Oncology Service, Hospital Universitario Araba, Vitoria-Gasteiz, Spain
| | - Maria Luque
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Idoia Morilla
- Breast Unit, Department of Medical Oncology, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Agostina Stradella
- Breast Unit, Department of Medical Oncology, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gemma Viñas
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain
| | - Javier Cortés
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Rovira i Virgili University, Reus, Spain
| | - Joan Brunet
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain.,Hereditary Cancer Programme, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain.,Hereditary Cancer Programme, Catalan Institute of Oncology (ICO), Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Eugeni López-Bonet
- Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona, Spain
| | - Margarita Garcia
- Clinical Research Unit, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Samiha Saidani
- Unit of Clinical Research, Catalan Institute of Oncology, Girona, Spain
| | | | | | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| |
Collapse
|
437
|
Abstract
For decades, dietary advice was based on the premise that high intakes of fat cause obesity, diabetes, heart disease, and possibly cancer. Recently, evidence for the adverse metabolic effects of processed carbohydrate has led to a resurgence in interest in lower-carbohydrate and ketogenic diets with high fat content. However, some argue that the relative quantity of dietary fat and carbohydrate has little relevance to health and that focus should instead be placed on which particular fat or carbohydrate sources are consumed. This review, by nutrition scientists with widely varying perspectives, summarizes existing evidence to identify areas of broad consensus amid ongoing controversy regarding macronutrients and chronic disease.
Collapse
Affiliation(s)
- David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Harvard Medical School, Boston, MA, USA.,Departments of Epidemiology and Nutrition, Harvard T. H. Chan School of Public Health and Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeff S Volek
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Marian L Neuhouser
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
438
|
Vallejo-Díaz J, Chagoyen M, Olazabal-Morán M, González-García A, Carrera AC. The Opposing Roles of PIK3R1/p85α and PIK3R2/p85β in Cancer. Trends Cancer 2019; 5:233-244. [PMID: 30961830 DOI: 10.1016/j.trecan.2019.02.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 01/04/2023]
Abstract
Dysregulation of the PI3K/PTEN pathway is a frequent event in cancer, and PIK3CA and PTEN are the most commonly mutated genes after TP53. PIK3R1 is the predominant regulatory isoform of PI3K. PIK3R2 is an ubiquitous isoform that has been so far overlooked, but data from The Cancer Genome Atlas shows that increased expression of PIK3R2 is also frequent in cancer. In contrast to PIK3R1, which is a tumor-suppressor gene, PIK3R2 is an oncogene. We review here the opposing roles of PIK3R1 and PIK3R2 in cancer, the regulatory mechanisms that control PIK3R2 expression, and emerging therapeutic approaches targeting PIK3R2.
Collapse
Affiliation(s)
- Jesús Vallejo-Díaz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Cantoblanco, Madrid E-28049, Spain
| | - Monica Chagoyen
- Department of Systems Biology, Centro Nacional de Biotecnología, CSIC, Universidad Autónoma de Madrid, Cantoblanco, Madrid E-28049, Spain
| | - Manuel Olazabal-Morán
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Cantoblanco, Madrid E-28049, Spain
| | - Ana González-García
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Cantoblanco, Madrid E-28049, Spain
| | - Ana Clara Carrera
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Cantoblanco, Madrid E-28049, Spain.
| |
Collapse
|
439
|
Kaklamani VG, Richardson AL, Arteaga CL. Exploring Biomarkers of Phosphoinositide 3-Kinase Pathway Activation in the Treatment of Hormone Receptor Positive, Human Epidermal Growth Receptor 2 Negative Advanced Breast Cancer. Oncologist 2019; 24:305-312. [PMID: 30651399 PMCID: PMC6519770 DOI: 10.1634/theoncologist.2018-0314] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/18/2018] [Indexed: 11/17/2022] Open
Abstract
Resistance to endocrine therapy (ET) is common in patients with hormone receptor positive (HR+) advanced breast cancer (ABC). Consequently, new targeted treatment options are needed in the post-ET setting, with validated biomarkers to inform treatment decisions. Hyperactivation of the phosphoinositide 3-kinase (PI3K) signaling pathway is common in ABC and is implicated in resistance to ET. The most frequent mechanism of PI3K pathway activation is activating mutations or amplification of PIK3CA, which encodes the α-isoform of the catalytic subunit of PI3K. Combining buparlisib, a pan-PI3K-targeted agent, with ET demonstrated modest clinical benefits in patients with aromatase inhibitor-resistant, HR+, human epidermal growth receptor 2 negative (HER2-) ABC in two phase III trials. Importantly, greater efficacy gains were observed in individuals with PIK3CA-mutated disease versus PIK3CA-wild-type tumors. Although the challenging safety profile did not support widespread use of this treatment combination, isoform-selective PI3K inhibitors may improve tolerability. In early clinical trials, promising disease control benefits were demonstrated with the PI3K isoform-selective inhibitors alpelisib and taselisib in patients with PIK3CA-mutated HR+, HER2- ABC. Ongoing biomarker-guided phase II/III studies may provide further opportunities to identify patients most likely to benefit from treatment with PI3K inhibitors and provide insight into optimizing the therapeutic index of PI3K inhibitors. Challenges facing the implementation of routine PIK3CA mutation testing must be addressed promptly so robust and reproducible genotyping can be obtained with liquid and tumor biopsies in a timely and cost-effective manner. IMPLICATIONS FOR PRACTICE: The development of phosphoinositide 3-kinase (PI3K) inhibitors, especially those that selectively target isoforms, may be an effective strategy for overcoming endocrine therapy resistance in hormone receptor positive, human epidermal growth receptor 2 negative advanced breast cancer. Early-phase studies have confirmed that patients with PIK3CA mutations respond best to PI3Kα-isoform inhibition. Ongoing phase III trials will provide further data regarding the efficacy and safety of PI3K inhibitors in patients with different biomarker profiles.
Collapse
Affiliation(s)
| | | | - Carlos L Arteaga
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
440
|
Abstract
PURPOSE OF REVIEW Altered glucose metabolism in cancer cells is an almost ubiquitous observation, yet hardly exploited therapeutically. However, ketogenic diets have gained growing attention in recent years as a nontoxic broad-spectrum approach to target this major metabolic difference between normal and cancer cells. Although much research still needs to be done, new knowledge has been gained about the optimal utilization of ketogenic diets for cancer treatment that this review aims to summarize. RECENT FINDINGS Although most preclinical studies indicate a therapeutic potential for ketogenic diets in cancer treatment, it is now becoming clear that not all tumors might respond positively. Early clinical trials have investigated ketogenic diets as a monotherapy and - while showing the safety of the approach even in advanced cancer patients - largely failed to prove survival prolonging effects. However, it gradually became clear that the greatest potential for ketogenic diets is as adjuvant treatments combined with pro-oxidative or targeted therapies initiated in early stages of the disease. Beneficial effects on body composition and quality of life have also been found. SUMMARY Ketogenic diets against cancer are worth further exploration, both in the laboratory and clinically. Patients wishing to undertake a ketogenic diet during therapy should receive dietary counselling to avoid common mistakes and optimize compliance. Future research should focus more on important clinical endpoints.
Collapse
Affiliation(s)
- Rainer Johannes Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Schweinfurt, Germany
| |
Collapse
|
441
|
Rauth M, Freund P, Orlova A, Grünert S, Tasic N, Han X, Ruan HB, Neubauer HA, Moriggl R. Cell Metabolism Control Through O-GlcNAcylation of STAT5: A Full or Empty Fuel Tank Makes a Big Difference for Cancer Cell Growth and Survival. Int J Mol Sci 2019; 20:E1028. [PMID: 30818760 PMCID: PMC6429193 DOI: 10.3390/ijms20051028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 12/23/2022] Open
Abstract
O-GlcNAcylation is a post-translational modification that influences tyrosine phosphorylation in healthy and malignant cells. O-GlcNAc is a product of the hexosamine biosynthetic pathway, a side pathway of glucose metabolism. It is essential for cell survival and proper gene regulation, mirroring the metabolic status of a cell. STAT3 and STAT5 proteins are essential transcription factors that can act in a mutational context-dependent manner as oncogenes or tumor suppressors. They regulate gene expression for vital processes such as cell differentiation, survival, or growth, and are also critically involved in metabolic control. The role of STAT3/5 proteins in metabolic processes is partly independent of their transcriptional regulatory role, but is still poorly understood. Interestingly, STAT3 and STAT5 are modified by O-GlcNAc in response to the metabolic status of the cell. Here, we discuss and summarize evidence of O-GlcNAcylation-regulating STAT function, focusing in particular on hyperactive STAT5A transplant studies in the hematopoietic system. We emphasize that a single O-GlcNAc modification is essential to promote development of neoplastic cell growth through enhancing STAT5A tyrosine phosphorylation. Inhibition of O-GlcNAcylation of STAT5A on threonine 92 lowers tyrosine phosphorylation of oncogenic STAT5A and ablates malignant transformation. We conclude on strategies for new therapeutic options to block O-GlcNAcylation in combination with tyrosine kinase inhibitors to target neoplastic cancer cell growth and survival.
Collapse
Affiliation(s)
- Manuel Rauth
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.
| | - Patricia Freund
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.
| | - Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.
- Ludwig Boltzmann Institute for Cancer Research, 1090 Vienna, Austria.
| | | | | | - Xiaonan Han
- Key Laboratory of Human Disease Comparative Medicine, the Ministry of Health, Institute of Laboratory Animal Sciences (ILAS), Beijing 100730, China.
- Chinese Academy of Medical Science (CAMS) and Peking Union Medical College (PUMC), Beijing 100006, China.
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH 45229-3026, USA.
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | - Heidi A Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.
- Ludwig Boltzmann Institute for Cancer Research, 1090 Vienna, Austria.
- Medical University Vienna, Vienna 1090, Austria.
| |
Collapse
|
442
|
Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int J Mol Sci 2019; 20:ijms20030676. [PMID: 30764482 PMCID: PMC6387440 DOI: 10.3390/ijms20030676] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
Systemic and local chronic inflammation might enhance the risk of pancreatic ductal adenocarcinoma (PDAC), and PDAC-associated inflammatory infiltrate in the tumor microenvironment concurs in enhancing tumor growth and metastasis. Inflammation is closely correlated with immunity, the same immune cell populations contributing to both inflammation and immune response. In the PDAC microenvironment, the inflammatory cell infiltrate is unbalanced towards an immunosuppressive phenotype, with a prevalence of myeloid derived suppressor cells (MDSC), M2 polarized macrophages, and Treg, over M1 macrophages, dendritic cells, and effector CD4⁺ and CD8⁺ T lymphocytes. The dynamic and continuously evolving cross-talk between inflammatory and cancer cells might be direct and contact-dependent, but it is mainly mediated by soluble and exosomes-carried cytokines. Among these, tumor necrosis factor alpha (TNFα) plays a relevant role in enhancing cancer risk, cancer growth, and cancer-associated cachexia. In this review, we describe the inflammatory cell types, the cytokines, and the mechanisms underlying PDAC risk, growth, and progression, with particular attention on TNFα, also in the light of the potential risks or benefits associated with anti-TNFα treatments.
Collapse
|
443
|
Wen PY, Touat M, Alexander BM, Mellinghoff IK, Ramkissoon S, McCluskey CS, Pelton K, Haidar S, Basu SS, Gaffey SC, Brown LE, Martinez-Ledesma JE, Wu S, Kim J, Wei W, Park MA, Huse JT, Kuhn JG, Rinne ML, Colman H, Agar NYR, Omuro AM, DeAngelis LM, Gilbert MR, de Groot JF, Cloughesy TF, Chi AS, Roberts TM, Zhao JJ, Lee EQ, Nayak L, Heath JR, Horky LL, Batchelor TT, Beroukhim R, Chang SM, Ligon AH, Dunn IF, Koul D, Young GS, Prados MD, Reardon DA, Yung WKA, Ligon KL. Buparlisib in Patients With Recurrent Glioblastoma Harboring Phosphatidylinositol 3-Kinase Pathway Activation: An Open-Label, Multicenter, Multi-Arm, Phase II Trial. J Clin Oncol 2019; 37:741-750. [PMID: 30715997 DOI: 10.1200/jco.18.01207] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Phosphatidylinositol 3-kinase (PI3K) signaling is highly active in glioblastomas. We assessed pharmacokinetics, pharmacodynamics, and efficacy of the pan-PI3K inhibitor buparlisib in patients with recurrent glioblastoma with PI3K pathway activation. METHODS This study was a multicenter, open-label, multi-arm, phase II trial in patients with PI3K pathway-activated glioblastoma at first or second recurrence. In cohort 1, patients scheduled for re-operation after progression received buparlisib for 7 to 13 days before surgery to evaluate brain penetration and modulation of the PI3K pathway in resected tumor tissue. In cohort 2, patients not eligible for re-operation received buparlisib until progression or unacceptable toxicity. Once daily oral buparlisib 100 mg was administered on a continuous 28-day schedule. Primary end points were PI3K pathway inhibition in tumor tissue and buparlisib pharmacokinetics in cohort 1 and 6-month progression-free survival (PFS6) in cohort 2. RESULTS Sixty-five patients were treated (cohort 1, n = 15; cohort 2, n = 50). In cohort 1, reduction of phosphorylated AKTS473 immunohistochemistry score was achieved in six (42.8%) of 14 patients, but effects on phosphoribosomal protein S6S235/236 and proliferation were not significant. Tumor-to-plasma drug level was 1.0. In cohort 2, four (8%) of 50 patients reached 6-month PFS6, and the median PFS was 1.7 months (95% CI, 1.4 to 1.8 months). The most common grade 3 or greater adverse events related to treatment were lipase elevation (n = 7 [10.8%]), fatigue (n = 4 [6.2%]), hyperglycemia (n = 3 [4.6%]), and elevated ALT (n = 3 [4.6%]). CONCLUSION Buparlisib had minimal single-agent efficacy in patients with PI3K-activated recurrent glioblastoma. Although buparlisib achieved significant brain penetration, the lack of clinical efficacy was explained by incomplete blockade of the PI3K pathway in tumor tissue. Integrative results suggest that additional study of PI3K inhibitors that achieve more-complete pathway inhibition may still be warranted.
Collapse
Affiliation(s)
- Patrick Y Wen
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Mehdi Touat
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Brian M Alexander
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | | | | | - Sam Haidar
- 1 Dana-Farber Cancer Institute, Boston, MA
| | - Sankha S Basu
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | | | - Shaofang Wu
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jungwoo Kim
- 5 California Institute of Technology, Pasadena, CA
| | - Wei Wei
- 6 David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,10 Institute for Systems Biology, Seattle, WA
| | - Mi-Ae Park
- 1 Dana-Farber Cancer Institute, Boston, MA
| | - Jason T Huse
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - John G Kuhn
- 7 The University of Texas, San Antonio, San Antonio, TX
| | - Mikael L Rinne
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Howard Colman
- 8 Huntsman Cancer Institute and University of Utah, Salt Lake City, UT
| | - Nathalie Y R Agar
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | - Mark R Gilbert
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - John F de Groot
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Timothy F Cloughesy
- 6 David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Andrew S Chi
- 9 New York University School of Medicine, New York, NY
| | | | | | - Eudocia Q Lee
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Lakshmi Nayak
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | | | | | | | - Rameen Beroukhim
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - Susan M Chang
- 12 University of California, San Francisco, San Francisco, CA
| | | | - Ian F Dunn
- 2 Brigham and Women's Hospital, Boston, MA
| | - Dimpy Koul
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | | | | | - David A Reardon
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| | - W K Alfred Yung
- 4 The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Keith L Ligon
- 1 Dana-Farber Cancer Institute, Boston, MA.,2 Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
444
|
Yehia L, Ngeow J, Eng C. PTEN-opathies: from biological insights to evidence-based precision medicine. J Clin Invest 2019; 129:452-464. [PMID: 30614812 DOI: 10.1172/jci121277] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The tumor suppressor phosphatase and tensin homolog (PTEN) classically counteracts the PI3K/AKT/mTOR signaling cascade. Germline pathogenic PTEN mutations cause PTEN hamartoma tumor syndrome (PHTS), featuring various benign and malignant tumors, as well as neurodevelopmental disorders such as autism spectrum disorder. Germline and somatic mosaic mutations in genes encoding components of the PI3K/AKT/mTOR pathway downstream of PTEN predispose to syndromes with partially overlapping clinical features, termed the "PTEN-opathies." Experimental models of PTEN pathway disruption uncover the molecular and cellular processes influencing clinical phenotypic manifestations. Such insights not only teach us about biological mechanisms in states of health and disease, but also enable more accurate gene-informed cancer risk assessment, medical management, and targeted therapeutics. Hence, the PTEN-opathies serve as a prototype for bedside to bench, and back to the bedside, practice of evidence-based precision medicine.
Collapse
Affiliation(s)
- Lamis Yehia
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joanne Ngeow
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre, Singapore.,Oncology Academic Program, Duke-NUS Graduate Medical School, Singapore
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Germline High Risk Cancer Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
445
|
Mirisola MG. Diet and Calorie Restriction. ENCYCLOPEDIA OF GERONTOLOGY AND POPULATION AGING 2019:1-10. [DOI: 10.1007/978-3-319-69892-2_123-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/02/2019] [Indexed: 09/02/2023]
|
446
|
Lamming DW, Bar-Peled L. Lysosome: The metabolic signaling hub. Traffic 2019; 20:27-38. [PMID: 30306667 PMCID: PMC6294686 DOI: 10.1111/tra.12617] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/17/2018] [Accepted: 10/07/2018] [Indexed: 12/27/2022]
Abstract
For the past five decades, the lysosome has been characterized as an unglamorous cellular recycling center. This notion has undergone a radical shift in the last 10 years, with new research revealing that this organelle serves as a major hub for metabolic signaling pathways. The discovery that master growth regulators, including the protein kinase mTOR (mechanistic target of rapamycin), make their home at the lysosomal surface has generated intense interest in the lysosome's key role in nutrient sensing and cellular homeostasis. The transcriptional networks required for lysosomal maintenance and function are just being unraveled and their connection to lysosome-based signaling pathways revealed. The catabolic and anabolic pathways that converge on the lysosome connect this organelle with multiple facets of cellular function; when these pathways are deregulated they underlie multiple human diseases, and promote cellular and organismal aging. Thus, understanding how lysosome-based signaling pathways function will not only illuminate the fascinating biology of this organelle but will also be critical in unlocking its therapeutic potentials.
Collapse
Affiliation(s)
- Dudley W. Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Liron Bar-Peled
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
447
|
Sarkar PL, Lee W, Williams ED, Lubik AA, Stylianou N, Shokoohmand A, Lehman ML, Hollier BG, Gunter JH, Nelson CC. Insulin Enhances Migration and Invasion in Prostate Cancer Cells by Up-Regulation of FOXC2. Front Endocrinol (Lausanne) 2019; 10:481. [PMID: 31379747 PMCID: PMC6652804 DOI: 10.3389/fendo.2019.00481] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/03/2019] [Indexed: 12/28/2022] Open
Abstract
Androgen deprivation therapy (ADT) is the standard treatment for advanced prostate cancer (PCa), yet many patients relapse with lethal metastatic disease. With this loss of androgens, increased cell plasticity has been observed as an adaptive response to ADT. This includes gain of invasive and migratory capabilities, which may contribute to PCa metastasis. Hyperinsulinemia, which develops as a side-effect of ADT, has been associated with increased tumor aggressiveness and faster treatment failure. We investigated the direct effects of insulin in PCa cells that may contribute to this progression. We measured cell migration and invasion induced by insulin using wound healing and transwell assays in a range of PCa cell lines of variable androgen dependency (LNCaP, 22RV1, DuCaP, and DU145 cell lines). To determine the molecular events driving insulin-induced invasion we used transcriptomics, quantitative real time-PCR, and immunoblotting in three PCa cell lines. Insulin increased invasiveness of PCa cells, upregulating Forkhead Box Protein C2 (FOXC2), and activating key PCa cell plasticity mechanisms including gene changes consistent with epithelial-to-mesenchymal transition (EMT) and a neuroendocrine phenotype. Additionally, analysis of publicly available clinical PCa tumor data showed metastatic prostate tumors demonstrate a positive correlation between insulin receptor expression and the EMT transcription factor FOXC2. The insulin receptor is not suitable to target clinically however, our data shows that actions of insulin in PCa cells may be suppressed by inhibiting downstream signaling molecules, PI3K and ERK1/2. This study identifies for the first time, a mechanism for insulin-driven cancer cell motility and supports the concept that targeting insulin signaling at the level of the PCa tumor may extend the therapeutic efficacy of ADT.
Collapse
Affiliation(s)
- Phoebe L. Sarkar
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| | - Wendy Lee
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| | - Elizabeth D. Williams
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| | - Amy A. Lubik
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Nataly Stylianou
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| | - Ali Shokoohmand
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| | - Melanie L. Lehman
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Brett G. Hollier
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| | - Jennifer H. Gunter
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
- *Correspondence: Jennifer H. Gunter
| | - Colleen C. Nelson
- Queensland University of Technology (QUT), Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
448
|
Goulden BD, Pacheco J, Dull A, Zewe JP, Deiters A, Hammond GRV. A high-avidity biosensor reveals plasma membrane PI(3,4)P 2 is predominantly a class I PI3K signaling product. J Cell Biol 2018; 218:1066-1079. [PMID: 30591513 PMCID: PMC6400549 DOI: 10.1083/jcb.201809026] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/20/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022] Open
Abstract
Class I phosphoinositide 3-OH kinase (PI3K) signaling is central to animal growth and metabolism, and pathological disruption of this pathway affects cancer and diabetes. However, the specific spatial/temporal dynamics and signaling roles of its minor lipid messenger, phosphatidylinositol (3,4)-bisphosphate (PI(3,4)P2), are not well understood. This owes principally to a lack of tools to study this scarce lipid. Here we developed a high-sensitivity genetically encoded biosensor for PI(3,4)P2, demonstrating high selectivity and specificity of the sensor for the lipid. We show that despite clear evidence for class II PI3K in PI(3,4)P2-driven function, the overwhelming majority of the lipid accumulates through degradation of class I PI3K-produced PIP3 However, we show that PI(3,4)P2 is also subject to hydrolysis by the tumor suppressor lipid phosphatase PTEN. Collectively, our results show that PI(3,4)P2 is potentially an important driver of class I PI3K-driven signaling and provides powerful new tools to begin to resolve the biological functions of this lipid downstream of class I and II PI3K.
Collapse
Affiliation(s)
- Brady D Goulden
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jonathan Pacheco
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Allyson Dull
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - James P Zewe
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
449
|
Park S, Kim YS, Kim DY, So I, Jeon JH. PI3K pathway in prostate cancer: All resistant roads lead to PI3K. Biochim Biophys Acta Rev Cancer 2018; 1870:198-206. [DOI: 10.1016/j.bbcan.2018.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
|
450
|
Affiliation(s)
- Marcus D Goncalves
- From the Meyer Cancer Center (M.D.G., B.D.H., L.C.C.) and the Division of Endocrinology (M.D.G.), Department of Medicine, Weill Cornell Medicine, New York
| | - Benjamin D Hopkins
- From the Meyer Cancer Center (M.D.G., B.D.H., L.C.C.) and the Division of Endocrinology (M.D.G.), Department of Medicine, Weill Cornell Medicine, New York
| | - Lewis C Cantley
- From the Meyer Cancer Center (M.D.G., B.D.H., L.C.C.) and the Division of Endocrinology (M.D.G.), Department of Medicine, Weill Cornell Medicine, New York
| |
Collapse
|