151
|
Yang R, Li SW, Chen Z, Zhou X, Ni W, Fu DA, Lu J, Kaye FJ, Wu L. Role of INSL4 Signaling in Sustaining the Growth and Viability of LKB1-Inactivated Lung Cancer. J Natl Cancer Inst 2020; 111:664-674. [PMID: 30423141 DOI: 10.1093/jnci/djy166] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 07/14/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The LKB1 tumor suppressor gene is commonly inactivated in non-small cell lung carcinomas (NSCLC), a major form of lung cancer. Targeted therapies for LKB1-inactivated lung cancer are currently unavailable. Identification of critical signaling components downstream of LKB1 inactivation has the potential to uncover rational therapeutic targets. Here we investigated the role of INSL4, a member of the insulin/IGF/relaxin superfamily, in LKB1-inactivated NSCLCs. METHODS INSL4 expression was analyzed using global transcriptome profiling, quantitative reverse transcription PCR, western blotting, enzyme-linked immunosorbent assay, and RNA in situ hybridization in human NSCLC cell lines and tumor specimens. INSL4 gene expression and clinical data from The Cancer Genome Atlas lung adenocarcinomas (n = 515) were analyzed using log-rank and Fisher exact tests. INSL4 functions were studied using short hairpin RNA (shRNA) knockdown, overexpression, transcriptome profiling, cell growth, and survival assays in vitro and in vivo. All statistical tests were two-sided. RESULTS INSL4 was identified as a novel downstream target of LKB1 deficiency and its expression was induced through aberrant CRTC-CREB activation. INSL4 was highly induced in LKB1-deficient NSCLC cells (up to 543-fold) and 9 of 41 primary tumors, although undetectable in all normal tissues except the placenta. Lung adenocarcinomas from The Cancer Genome Atlas with high and low INSL4 expression (with the top 10th percentile as cutoff) showed statistically significant differences for advanced tumor stage (P < .001), lymph node metastasis (P = .001), and tumor size (P = .01). The INSL4-high group showed worse survival than the INSL4-low group (P < .001). Sustained INSL4 expression was required for the growth and viability of LKB1-inactivated NSCLC cells in vitro and in a mouse xenograft model (n = 5 mice per group). Expression profiling revealed INSL4 as a critical regulator of cell cycle, growth, and survival. CONCLUSIONS LKB1 deficiency induces an autocrine INSL4 signaling that critically supports the growth and survival of lung cancer cells. Therefore, aberrant INSL4 signaling is a promising therapeutic target for LKB1-deficient lung cancers.
Collapse
Affiliation(s)
- Rongqiang Yang
- Department of Molecular Genetics and Microbiology.,UF Health Cancer Center
| | - Steven W Li
- Department of Molecular Genetics and Microbiology.,UF Health Cancer Center.,Department of Medicine
| | - Zirong Chen
- Department of Molecular Genetics and Microbiology.,UF Health Cancer Center
| | - Xin Zhou
- Department of Molecular Genetics and Microbiology.,UF Health Cancer Center
| | - Wei Ni
- Department of Molecular Genetics and Microbiology.,UF Genetics Institute
| | - Dongtao A Fu
- Department of Pathology, Immunology and Laboratory Medicine
| | - Jianrong Lu
- Department of Molecular Genetics and Microbiology.,UF Health Cancer Center.,Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL
| | | | - Lizi Wu
- Department of Molecular Genetics and Microbiology.,UF Health Cancer Center.,UF Genetics Institute
| |
Collapse
|
152
|
Li F, Simon MC. Cancer Cells Don't Live Alone: Metabolic Communication within Tumor Microenvironments. Dev Cell 2020; 54:183-195. [PMID: 32640203 DOI: 10.1016/j.devcel.2020.06.018] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/10/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023]
Abstract
Solid tumors reside in harsh tumor microenvironments (TMEs) together with various stromal cell types. During tumor progression and metastasis, both tumor and stromal cells undergo rapid metabolic adaptations. Tumor cells metabolically coordinate or compete with their "neighbors" to maintain biosynthetic and bioenergetic demands while escaping immunosurveillance or therapeutic interventions. Here, we provide an update on metabolic communication between tumor cells and heterogeneous stromal components in primary and metastatic TMEs and discuss emerging strategies to target metabolic communications for improved cancer treatments.
Collapse
Affiliation(s)
- Fuming Li
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
153
|
Zhu X, Chen HH, Gao CY, Zhang XX, Jiang JX, Zhang Y, Fang J, Zhao F, Chen ZG. Energy metabolism in cancer stem cells. World J Stem Cells 2020; 12:448-461. [PMID: 32742562 PMCID: PMC7360992 DOI: 10.4252/wjsc.v12.i6.448] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/09/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
Normal cells mainly rely on oxidative phosphorylation as an effective energy source in the presence of oxygen. In contrast, most cancer cells use less efficient glycolysis to produce ATP and essential biomolecules. Cancer cells gain the characteristics of metabolic adaptation by reprogramming their metabolic mechanisms to meet the needs of rapid tumor growth. A subset of cancer cells with stem characteristics and the ability to regenerate exist throughout the tumor and are therefore called cancer stem cells (CSCs). New evidence indicates that CSCs have different metabolic phenotypes compared with differentiated cancer cells. CSCs can dynamically transform their metabolic state to favor glycolysis or oxidative metabolism. The mechanism of the metabolic plasticity of CSCs has not been fully elucidated, and existing evidence indicates that the metabolic phenotype of cancer cells is closely related to the tumor microenvironment. Targeting CSC metabolism may provide new and effective methods for the treatment of tumors. In this review, we summarize the metabolic characteristics of cancer cells and CSCs and the mechanisms of the metabolic interplay between the tumor microenvironment and CSCs, and discuss the clinical implications of targeting CSC metabolism.
Collapse
Affiliation(s)
- Xuan Zhu
- Department of Radiation Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Hui-Hui Chen
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Chen-Yi Gao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Xin-Xin Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Jing-Xin Jiang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Yi Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Jun Fang
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Feng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Zhi-Gang Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China.
| |
Collapse
|
154
|
Metformin: (future) best friend of the radiation oncologist? Radiother Oncol 2020; 151:95-105. [PMID: 32592892 DOI: 10.1016/j.radonc.2020.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 02/08/2023]
Abstract
Several molecules are being investigated for their ability to enhance the anti-tumor effect of radiotherapy. The widely prescribed antidiabetic drug metformin has been suggested to possess anti-cancer activity; data indicate that metformin could also enhance radiation sensitivity. The purpose of this review is to summarize current knowledge on the specific effect of metformin in the field of RT, while also discussing the many unknowns that persist. Preclinical models point to multiple mechanisms involved in the radiosensitizing effects of metformin that are mainly linked to mitochondrial complex I inhibition and AMP-activated protein kinase. Transposition of results from bench to bedside will be discussed through the lens of the drug concentration, its potential limits in human settings, and possible alternatives. Clinical data suggest metformin improves progression-free and overall survival in patients for many different cancers treated with RT; nevertheless, the results are not always consistent. The main limitations of the reviewed literature are the retrospective nature of studies, and most of the time, a lack of information on MTF treatment duration and the administered dosages. Despite these limitations, the possible mechanisms of the role of metformin and its utility in enhancing radiotherapy treatments are analyzed. Ongoing clinical trials are also discussed.
Collapse
|
155
|
Kery M, Papandreou I. Emerging strategies to target cancer metabolism and improve radiation therapy outcomes. Br J Radiol 2020; 93:20200067. [PMID: 32462882 DOI: 10.1259/bjr.20200067] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer-specific metabolic changes support the anabolic needs of the rapidly growing tumor, maintain a favorable redox balance, and help cells adapt to microenvironmental stresses like hypoxia and nutrient deprivation. Radiation is extensively applied in a large number of cancer treatment protocols but despite its curative potential, radiation resistance and treatment failures pose a serious problem. Metabolic control of DNA integrity and genomic stability can occur through multiple processes, encompassing cell cycle regulation, nucleotide synthesis, epigenetic regulation of gene activity, and antioxidant defenses. Given the important role of metabolic pathways in oxidative damage responses, it is necessary to assess the potential for tumor-specific radiosensitization by novel metabolism-targeted therapies. Additionally, there are opportunities to identify molecular and functional biomarkers of vulnerabilities to combination treatments, which could then inform clinical decisions. Here, we present a curated list of metabolic pathways in the context of ionizing radiation responses. Glutamine metabolism influences DNA damage responses by mechanisms such as synthesis of nucleotides for DNA repair or of glutathione for ROS detoxification. Repurposed oxygen consumption inhibitors have shown promising radiosensitizing activity against murine model tumors and are now in clinical trials. Production of 2-hydroxy glutarate by isocitrate dehydrogenase1/2 neomorphic oncogenic mutants interferes with the function of α-ketoglutarate-dependent enzymes and modulates Ataxia Telangiectasia Mutated (ATM) signaling and glutathione pools. Radiation-induced oxidative damage to membrane phospholipids promotes ferroptotic cell loss and cooperates with immunotherapies to improve tumor control. In summary, there are opportunities to enhance the efficacy of radiotherapy by exploiting cell-inherent vulnerabilities and dynamic microenvironmental components of the tumor.
Collapse
Affiliation(s)
| | - Ioanna Papandreou
- Department of Radiation Oncology, Wexner Medical Center and Comprehensive Cancer Center The Ohio State University Columbus, OH, USA
| |
Collapse
|
156
|
Uras IZ, Moll HP, Casanova E. Targeting KRAS Mutant Non-Small-Cell Lung Cancer: Past, Present and Future. Int J Mol Sci 2020; 21:E4325. [PMID: 32560574 PMCID: PMC7352653 DOI: 10.3390/ijms21124325] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the most frequent cancer with an aggressive clinical course and high mortality rates. Most cases are diagnosed at advanced stages when treatment options are limited and the efficacy of chemotherapy is poor. The disease has a complex and heterogeneous background with non-small-cell lung cancer (NSCLC) accounting for 85% of patients and lung adenocarcinoma being the most common histological subtype. Almost 30% of adenocarcinomas of the lung are driven by an activating Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation. The ability to inhibit the oncogenic KRAS has been the holy grail of cancer research and the search for inhibitors is immensely ongoing as KRAS-mutated tumors are among the most aggressive and refractory to treatment. Therapeutic strategies tailored for KRAS+ NSCLC rely on the blockage of KRAS functional output, cellular dependencies, metabolic features, KRAS membrane associations, direct targeting of KRAS and immunotherapy. In this review, we provide an update on the most recent advances in anti-KRAS therapy for lung tumors with mechanistic insights into biological diversity and potential clinical implications.
Collapse
Affiliation(s)
- Iris Z. Uras
- Department of Pharmacology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Herwig P. Moll
- Department of Physiology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria; (H.P.M.); (E.C.)
| | - Emilio Casanova
- Department of Physiology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria; (H.P.M.); (E.C.)
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), 1090 Vienna, Austria
| |
Collapse
|
157
|
Shang C, Zhou H, Liu W, Shen T, Luo Y, Huang S. Iron chelation inhibits mTORC1 signaling involving activation of AMPK and REDD1/Bnip3 pathways. Oncogene 2020; 39:5201-5213. [PMID: 32541839 PMCID: PMC7366895 DOI: 10.1038/s41388-020-1366-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022]
Abstract
The mammalian target of rapamycin (mTOR) functions as two complexes (mTORC1 and mTORC2), regulating cell growth and metabolism. Aberrant mTOR signaling occurs frequently in cancers, so mTOR has become an attractive target for cancer therapy. Iron chelators have emerged as promising anticancer agents. However, the mechanisms underlying the anticancer action of iron chelation are not fully understood. Particularly, reports on the effects of iron chelation on mTOR complexes are inconsistent or controversial. Here, we found that iron chelators consistently inhibited mTORC1 signaling, which was blocked by pretreatment with ferrous sulfate. Mechanistically, iron chelation-induced mTORC1 inhibition was not related to ROS induction, copper chelation, or PP2A activation. Instead, activation of AMPK pathway mainly and activation of both HIF-1/REDD1 and Bnip3 pathways partially contribute to iron chelation-induced mTORC1 inhibition. Our findings indicate that iron chelation inhibits mTORC1 via multiple pathways and iron is essential for mTORC1 activation.
Collapse
Affiliation(s)
- Chaowei Shang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| | - Hongyu Zhou
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| | - Wang Liu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| | - Tao Shen
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA. .,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.
| |
Collapse
|
158
|
Santarpia M, Aguilar A, Chaib I, Cardona AF, Fancelli S, Laguia F, Bracht JWP, Cao P, Molina-Vila MA, Karachaliou N, Rosell R. Non-Small-Cell Lung Cancer Signaling Pathways, Metabolism, and PD-1/PD-L1 Antibodies. Cancers (Basel) 2020; 12:E1475. [PMID: 32516941 PMCID: PMC7352732 DOI: 10.3390/cancers12061475] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 12/16/2022] Open
Abstract
Treatment of advanced (metastatic) non-small-cell lung cancer (NSCLC) is currently mainly based on immunotherapy with antibodies against PD-1 or PD-L1, alone, or in combination with chemotherapy. In locally advanced NSCLC and in early resected stages, immunotherapy is also employed. Tumor PD-L1 expression by immunohistochemistry is considered the standard practice. Response rate is low, with median progression free survival very short in the vast majority of studies reported. Herein, numerous biological facets of NSCLC are described involving driver genetic lesions, mutations ad fusions, PD-L1 glycosylation, ferroptosis and metabolic rewiring in NSCLC and lung adenocarcinoma (LUAD). Novel concepts, such as immune-transmitters and the effect of neurotransmitters in immune evasion and tumor growth, the nascent relevance of necroptosis and pyroptosis, possible new biomarkers, such as gasdermin D and gasdermin E, the conundrum of K-Ras mutations in LUADs, with the growing recognition of liver kinase B1 (LKB1) and metabolic pathways, including others, are also commented. The review serves to charter diverse treatment solutions, depending on the main altered signaling pathways, in order to have effectual immunotherapy. Tumor PDCD1 gene (encoding PD-1) has been recently described, in equilibrium with tumor PD-L1 (encoded by PDCD1LG1). Such description explains tumor hyper-progression, which has been reported in several studies, and poises the fundamental criterion that IHC PD-L1 expression as a biomarker should be revisited.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Department of Human Pathology “G. Barresi”, Medical Oncology Unit, University of Messina, 98122 Messina, Italy;
| | - Andrés Aguilar
- Instituto Oncológico Dr Rosell, Hospital Universitario Quirón-Dexeus, 08028 Barcelona, Spain;
| | - Imane Chaib
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916 Badalona, Spain; (I.C.); (S.F.); (F.L.)
| | - Andrés Felipe Cardona
- Foundation for Clinical and Applied Cancer Research-FICMAC Translational Oncology, Bogotá 100110, Colombia;
| | - Sara Fancelli
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916 Badalona, Spain; (I.C.); (S.F.); (F.L.)
| | - Fernando Laguia
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916 Badalona, Spain; (I.C.); (S.F.); (F.L.)
| | | | - Peng Cao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Miguel Angel Molina-Vila
- Pangaea Oncology, Hospital Universitario Quirón-Dexeus, 08028 Barcelona, Spain; (J.W.P.B.); (M.A.M.-V.)
| | | | - Rafael Rosell
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916 Badalona, Spain; (I.C.); (S.F.); (F.L.)
| |
Collapse
|
159
|
Brown JR, Chan DK, Shank JJ, Griffith KA, Fan H, Szulawski R, Yang K, Reynolds RK, Johnston C, McLean K, Uppal S, Liu JR, Cabrera L, Taylor SE, Orr BC, Modugno F, Mehta P, Bregenzer M, Mehta G, Shen H, Coffman LG, Buckanovich RJ. Phase II clinical trial of metformin as a cancer stem cell-targeting agent in ovarian cancer. JCI Insight 2020; 5:133247. [PMID: 32369446 PMCID: PMC7308054 DOI: 10.1172/jci.insight.133247] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUNDEpidemiologic studies suggest that metformin has antitumor effects. Laboratory studies indicate metformin impacts cancer stem-like cells (CSCs). As part of a phase II trial, we evaluated the impact of metformin on CSC number and on carcinoma-associated mesenchymal stem cells (CA-MSCs) and clinical outcomes in nondiabetic patients with advanced-stage epithelial ovarian cancer (EOC).METHODSThirty-eight patients with stage IIC (n = 1)/III (n = 25)/IV (n = 12) EOC were treated with either (a) neoadjuvant metformin, debulking surgery, and adjuvant chemotherapy plus metformin or (b) neoadjuvant chemotherapy and metformin, interval debulking surgery, and adjuvant chemotherapy plus metformin. Metformin-treated tumors, compared with historical controls, were evaluated for CSC number and chemotherapy response. Primary endpoints were (a) a 2-fold or greater reduction in aldehyde dehydrogenase-positive (ALDH+) CD133+ CSCs and (b) a relapse-free survival at 18 months of more than 50%.RESULTSMetformin was well tolerated. Median progression-free survival was 18.0 months (95% CI 14.0-21.6) with relapse-free survival at 18 months of 59.3% (95% CI 38.6-70.5). Median overall survival was 57.9 months (95% CI 28.0-not estimable). Tumors treated with metformin had a 2.4-fold decrease in ALDH+CD133+ CSCs and increased sensitivity to cisplatin ex vivo. Furthermore, metformin altered the methylation signature in CA-MSCs, which prevented CA-MSC-driven chemoresistance in vitro.CONCLUSIONTranslational studies confirm an impact of metformin on EOC CSCs and suggest epigenetic change in the tumor stroma may drive the platinum sensitivity ex vivo. Consistent with this, metformin therapy was associated with better-than-expected overall survival, supporting the use of metformin in phase III studies.TRIAL REGISTRATIONClinicalTrials.gov NCT01579812.
Collapse
Affiliation(s)
- Jason R. Brown
- Division of Hematology and Oncology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Daniel K. Chan
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jessica J. Shank
- Department of Obstetrics and Gynecology, Naval Medical Center, San Diego, California, USA
| | - Kent A. Griffith
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Huihui Fan
- Van Andel Institute, Grand Rapids, Michigan, USA
| | - Robert Szulawski
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Kun Yang
- Division of Hematology and Oncology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - R. Kevin Reynolds
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Carolyn Johnston
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Karen McLean
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Shitanshu Uppal
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - J. Rebecca Liu
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Lourdes Cabrera
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Sarah E. Taylor
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Brian C. Orr
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Francesmary Modugno
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Pooja Mehta
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Michael Bregenzer
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Geeta Mehta
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Hui Shen
- Van Andel Institute, Grand Rapids, Michigan, USA
| | - Lan G. Coffman
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Ronald J. Buckanovich
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
160
|
Izreig S, Gariepy A, Kaymak I, Bridges HR, Donayo AO, Bridon G, DeCamp LM, Kitchen-Goosen SM, Avizonis D, Sheldon RD, Laister RC, Minden MD, Johnson NA, Duchaine TF, Rudoltz MS, Yoo S, Pollak MN, Williams KS, Jones RG. Repression of LKB1 by miR-17∼92 Sensitizes MYC-Dependent Lymphoma to Biguanide Treatment. Cell Rep Med 2020; 1:100014. [PMID: 32478334 PMCID: PMC7249503 DOI: 10.1016/j.xcrm.2020.100014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/04/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022]
Abstract
Cancer cells display metabolic plasticity to survive stresses in the tumor microenvironment. Cellular adaptation to energetic stress is coordinated in part by signaling through the liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) pathway. Here, we demonstrate that miRNA-mediated silencing of LKB1 confers sensitivity of lymphoma cells to mitochondrial inhibition by biguanides. Using both classic (phenformin) and newly developed (IM156) biguanides, we demonstrate that elevated miR-17∼92 expression in Myc+ lymphoma cells promotes increased apoptosis to biguanide treatment in vitro and in vivo. This effect is driven by the miR-17-dependent silencing of LKB1, which reduces AMPK activation in response to complex I inhibition. Mechanistically, biguanide treatment induces metabolic stress in Myc+ lymphoma cells by inhibiting TCA cycle metabolism and mitochondrial respiration, exposing metabolic vulnerability. Finally, we demonstrate a direct correlation between miR-17∼92 expression and biguanide sensitivity in human cancer cells. Our results identify miR-17∼92 expression as a potential biomarker for biguanide sensitivity in malignancies.
Collapse
Affiliation(s)
- Said Izreig
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Alexandra Gariepy
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Irem Kaymak
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Hannah R. Bridges
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Ariel O. Donayo
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Gaëlle Bridon
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Metabolomics Core Facility, McGill University, Montreal, QC H3A 1A3, Canada
| | - Lisa M. DeCamp
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Susan M. Kitchen-Goosen
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Daina Avizonis
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Metabolomics Core Facility, McGill University, Montreal, QC H3A 1A3, Canada
| | - Ryan D. Sheldon
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Rob C. Laister
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, Toronto, ON M5G 2M9, Canada
| | - Mark D. Minden
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Nathalie A. Johnson
- Lady Davis Institute of the Jewish General Hospital and Department of Oncology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Thomas F. Duchaine
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | | | - Sanghee Yoo
- ImmunoMet Therapeutics, Houston, TX 77021, USA
| | - Michael N. Pollak
- Lady Davis Institute of the Jewish General Hospital and Department of Oncology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Kelsey S. Williams
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Russell G. Jones
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
161
|
Vernieri C, Ganzinelli M, Rulli E, Farina G, Bettini AC, Bareggi C, Rosso L, Signorelli D, Galli G, Lo Russo G, Proto C, Moro M, Indraccolo S, Busico A, Sozzi G, Torri V, Marabese M, Massimo B, Garassino MC. LKB1 mutations are not associated with the efficacy of first-line and second-line chemotherapy in patients with advanced non-small-cell lung cancer (NSCLC): a post hoc analysis of the TAILOR trial. ESMO Open 2020; 5:e000748. [PMID: 32467099 PMCID: PMC7259832 DOI: 10.1136/esmoopen-2020-000748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE In patients with advanced lung adenocarcinoma, the impact of LKB1 mutations on cytotoxic chemotherapy efficacy remains poorly explored. Here, we aimed at investigating the potential impact of LKB1 mutational status on chemotherapy efficacy in advanced non-small-cell lung cancer (NSCLC) patients enrolled in the TArceva Italian Lung Optimisation tRial (TAILOR) trial. METHODS The multicenter TAILOR trial randomised patients with EGFR-wild type (wt) advanced NSCLC progressing on/after previous platinum-based chemotherapy to receive docetaxel or erlotinib. Here, we evaluated the impact of LKB1 mutational status on progression-free survival (PFS) and overall survival (OS) in patients treated with second-line docetaxel/erlotinib or during prior platinum-based chemotherapy. RESULTS Out of 222 patients randomised in the TAILOR trial, left-over tumour tissues were available for 188 patients, and 120 patients with evaluable LKB1 status were included. Of them, 17 (14.17%) patients had LKB1-mutated tumours, while 103 (85.83%) had LKB1-wt disease. During second-line treatment, PFS and OS were not statistically significantly different in patients with LKB1-mutated when compared with LKB1-wt NSCLC (adjusted HR (aHR)=1.29, 95% CI 0.75 to 2.21; p=0.364 and aHR=1.41, 95% CI 0.82 to 2.44; p=0.218, respectively). Similarly, we found no significant association between LKB1 mutations and patient PFS or OS during prior first-line platinum-based chemotherapy (aHR=1.04, 95% CI 0.55 to 1.97; p=0.910 and aHR=0.83, 95% CI 0.42 to 1.65; p=0.602, respectively). CONCLUSION Among advanced NSCLC patients receiving two lines of systemic therapy, LKB1 mutations were not associated with PFS or OS during second-line docetaxel or prior first-line platinum-based chemotherapy. While larger prospective trials are needed to confirm our findings, cytotoxic chemotherapy remains the backbone of investigational combination strategies in this patient population.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy; IFOM, the FIRC Institute of Molecular Oncology, Milano, Italy.
| | - Monica Ganzinelli
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Eliana Rulli
- Laboratory of Methodology for Clinical Research, Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri Sede di Milano, Milano, Lombardia, Italy
| | - Gabriella Farina
- Department of Oncology, ASST Fatebenefratelli Sacco, Milano, Lombardia, Italy
| | | | - Claudia Bareggi
- Oncology Unit, La Fondazione IRCCS Ca' Granda Ospedale Maggiore di Milano Policlinico, Milano, Lombardia, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplant Unit, La Fondazione IRCCS Ca' Granda Ospedale Maggiore di Milano Policlinico, Milano, Lombardia, Italy
| | - Diego Signorelli
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Giulia Galli
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Giuseppe Lo Russo
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Claudia Proto
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Massimo Moro
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Stefano Indraccolo
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Veneto, Italy
| | - Adele Busico
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Gabriella Sozzi
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Valter Torri
- Laboratory of Methodology for Clinical Research, Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri Sede di Milano, Milano, Lombardia, Italy
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri Sede di Milano, Milano, Lombardia, Italy
| | - Broggini Massimo
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri Sede di Milano, Milano, Lombardia, Italy
| | - Marina C Garassino
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| |
Collapse
|
162
|
Faubert B, Solmonson A, DeBerardinis RJ. Metabolic reprogramming and cancer progression. Science 2020; 368:368/6487/eaaw5473. [PMID: 32273439 DOI: 10.1126/science.aaw5473] [Citation(s) in RCA: 1390] [Impact Index Per Article: 278.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
Metabolic reprogramming is a hallmark of malignancy. As our understanding of the complexity of tumor biology increases, so does our appreciation of the complexity of tumor metabolism. Metabolic heterogeneity among human tumors poses a challenge to developing therapies that exploit metabolic vulnerabilities. Recent work also demonstrates that the metabolic properties and preferences of a tumor change during cancer progression. This produces distinct sets of vulnerabilities between primary tumors and metastatic cancer, even in the same patient or experimental model. We review emerging concepts about metabolic reprogramming in cancer, with particular attention on why metabolic properties evolve during cancer progression and how this information might be used to develop better therapeutic strategies.
Collapse
Affiliation(s)
- Brandon Faubert
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ashley Solmonson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA. .,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
163
|
Tumor Milieu Controlled by RB Tumor Suppressor. Int J Mol Sci 2020; 21:ijms21072450. [PMID: 32244804 PMCID: PMC7177274 DOI: 10.3390/ijms21072450] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 03/31/2020] [Indexed: 02/08/2023] Open
Abstract
The RB gene is one of the most frequently mutated genes in human cancers. Canonically, RB exerts its tumor suppressive activity through the regulation of the G1/S transition during cell cycle progression by modulating the activity of E2F transcription factors. However, aberration of the RB gene is most commonly detected in tumors when they gain more aggressive phenotypes, including metastatic activity or drug resistance, rather than accelerated proliferation. This implicates RB controls' malignant progression to a considerable extent in a cell cycle-independent manner. In this review, we highlight the multifaceted functions of the RB protein in controlling tumor lineage plasticity, metabolism, and the tumor microenvironment (TME), with a focus on the mechanism whereby RB controls the TME. In brief, RB inactivation in several types of cancer cells enhances production of pro-inflammatory cytokines, including CCL2, through upregulation of mitochondrial reactive oxygen species (ROS) production. These factors not only accelerate the growth of cancer cells in a cell-autonomous manner, but also stimulate non-malignant cells in the TME to generate a pro-tumorigenic niche in a non-cell-autonomous manner. Here, we discuss the biological and pathological significance of the non-cell-autonomous functions of RB and attempt to predict their potential clinical relevance to cancer immunotherapy.
Collapse
|
164
|
Abstract
Lung cancer is a heterogeneous genomic disease. Smoking remains the primary cause. Genetic susceptibility and environmental exposures are responsible for 10% to 15% of cases. Targeted therapies improve survival in patients with tumors with oncogenic drivers. It is critical to expand our understanding of genetic alterations in non-small cell lung cancer to increase the available targeted therapies. Alterations beyond epidermal growth factor receptor (EGFR), ALK, and ROS1 exemplify lung cancer's complexity and the need for investments in precision therapy to extend patient survival and improve outcomes. This article covers genetic targets beyond EGFR, ALK and ROS1, their novel agents, challenges, and future directions.
Collapse
Affiliation(s)
- Karen L Reckamp
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| |
Collapse
|
165
|
Chen K, Li Y, Guo Z, Zeng Y, Zhang W, Wang H. Metformin: current clinical applications in nondiabetic patients with cancer. Aging (Albany NY) 2020; 12:3993-4009. [PMID: 32074084 PMCID: PMC7066888 DOI: 10.18632/aging.102787] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/19/2020] [Indexed: 02/06/2023]
Abstract
Metformin is one of the most commonly used first-line oral medications for type 2 diabetes mellitus. Multiple observational studies, reviewed in numerous systematic reviews, have shown that metformin treatment may not only reduce the risk of cancer but may also improve the efficacy of cancer treatment in diabetic patients. Recent studies have been conducted to determine whether a similar protective effect can be demonstrated in nondiabetic cancer patients. However, the results are controversial. The potential optimal dose, schedule, and duration of metformin treatment and the heterogeneity of histological subtypes and genotypes among cancer patients might contribute to the different clinical benefits. In addition, as the immune property of metformin was investigated, further studies of the immunomodulatory effect of metformin on cancer cells should also be taken into account to optimize its clinical use. In this review, we present and discuss the latest findings regarding the anticancer potential of metformin in nondiabetic patients with cancer.
Collapse
Affiliation(s)
- Kailin Chen
- Key Laboratory of Translational Radiation Oncology, Hunan Province, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, P.R. China
| | - Yajun Li
- Department of Lymphoma and Hematology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, P.R. China
| | - Zhen Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, P.R. China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Yong Zeng
- Translational Medicine Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, P.R. China.,Engineering Technology Research Center for Diagnosis-Treatment and Application of Tumor Liquid Biopsy, Changsha 410013, Hunan, P.R. China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, P.R. China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Hui Wang
- Key Laboratory of Translational Radiation Oncology, Hunan Province, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, P.R. China
| |
Collapse
|
166
|
Ngoi NYL, Eu JQ, Hirpara J, Wang L, Lim JSJ, Lee SC, Lim YC, Pervaiz S, Goh BC, Wong ALA. Targeting Cell Metabolism as Cancer Therapy. Antioxid Redox Signal 2020; 32:285-308. [PMID: 31841375 DOI: 10.1089/ars.2019.7947] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Cancer cells exhibit altered metabolic pathways to keep up with biosynthetic and reduction-oxidation needs during tumor proliferation and metastasis. The common induction of metabolic pathways during cancer progression, regardless of cancer histio- or genotype, makes cancer metabolism an attractive target for therapeutic exploitation. Recent Advances: Emerging data suggest that these altered pathways may even result in resistance to anticancer therapies. Identifying specific metabolic dependencies that are unique to cancer cells has proved challenging in this field, limiting the therapeutic window for many candidate drug approaches. Critical Issues: Cancer cells display significant metabolic flexibility in nutrient-limited environments, hampering the longevity of suppressing cancer metabolism through any singular approach. Combinatorial "synthetic lethal" approaches may have a better chance for success and promising strategies are reviewed here. The dynamism of the immune system adds a level of complexity, as various immune populations in the tumor microenvironment often share metabolic pathways with cancer, with successive alterations during immune activation and quiescence. Decoding the reprogramming of metabolic pathways within cancer cells and stem cells, as well as examining metabolic symbiosis between components of the tumor microenvironment, would be essential to further meaningful drug development within the tumor's metabolic ecosystem. Future Directions: In this article, we examine evidence for the therapeutic potential of targeting metabolic alterations in cancer, and we discuss the drawbacks and successes that have stimulated this field.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Jie Qing Eu
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Jayshree Hirpara
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Joline S J Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Yaw-Chyn Lim
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Andrea L A Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| |
Collapse
|
167
|
Steinberg GR, Carling D. AMP-activated protein kinase: the current landscape for drug development. Nat Rev Drug Discov 2020; 18:527-551. [PMID: 30867601 DOI: 10.1038/s41573-019-0019-2] [Citation(s) in RCA: 472] [Impact Index Per Article: 94.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the discovery of AMP-activated protein kinase (AMPK) as a central regulator of energy homeostasis, many exciting insights into its structure, regulation and physiological roles have been revealed. While exercise, caloric restriction, metformin and many natural products increase AMPK activity and exert a multitude of health benefits, developing direct activators of AMPK to elicit beneficial effects has been challenging. However, in recent years, direct AMPK activators have been identified and tested in preclinical models, and a small number have entered clinical trials. Despite these advances, which disease(s) represent the best indications for therapeutic AMPK activation and the long-term safety of such approaches remain to be established.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - David Carling
- Cellular Stress Group, Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, Imperial College, London, UK
| |
Collapse
|
168
|
Thomas HE, Zhang Y, Stefely JA, Veiga SR, Thomas G, Kozma SC, Mercer CA. Mitochondrial Complex I Activity Is Required for Maximal Autophagy. Cell Rep 2020; 24:2404-2417.e8. [PMID: 30157433 PMCID: PMC6298213 DOI: 10.1016/j.celrep.2018.07.101] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/17/2018] [Accepted: 07/30/2018] [Indexed: 01/01/2023] Open
Abstract
Cells adapt to nutrient and energy deprivation by inducing autophagy, which is regulated by the mammalian target of rapamycin (mTOR) and AMP-activated protein kinases (AMPKs). We found that cell metabolism significantly influences the ability to induce autophagy, with mitochondrial complex I function being an important factor in the initiation, amplitude, and duration of the response. We show that phenformin or genetic defects in complex I suppressed autophagy induced by mTOR inhibitors, whereas autophagy was enhanced by strategies that increased mitochondrial metabolism. We report that mTOR inhibitors significantly increased select phospholipids and mitochondrial-associated membranes (MAMs) in a complex I-dependent manner. We attribute the complex I autophagy defect to the inability to increase MAMs, limiting phosphatidylserine decarboxylase (PISD) activity and mitochondrial phosphatidylethanolamine (mtPE), which support autophagy. Our data reveal the dynamic and metabolic regulation of autophagy.
Collapse
Affiliation(s)
- Hala Elnakat Thomas
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Yu Zhang
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Jonathan A Stefely
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA; Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sonia R Veiga
- Laboratory of Metabolism and Cancer, Catalan Institute of Oncology, ICO, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - George Thomas
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA; Laboratory of Metabolism and Cancer, Catalan Institute of Oncology, ICO, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain; Unit de Biochemistry, Department of Physiological Sciences II, Faculty of Medicine, Campus Universitari de Bellvitge-IDIBELL, University of Barcelona, 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Sara C Kozma
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA; Laboratory of Metabolism and Cancer, Catalan Institute of Oncology, ICO, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Carol A Mercer
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
169
|
Inhibition of mTORC1/P70S6K pathway by Metformin synergistically sensitizes Acute Myeloid Leukemia to Ara-C. Life Sci 2020; 243:117276. [PMID: 31926250 DOI: 10.1016/j.lfs.2020.117276] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/24/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022]
Abstract
AIMS Chemo-resistance still was the main obstacle for AML patients, more effective and less toxic forms of therapies were desperately needed. Metformin, a classic hypoglycemic drug for diabetes recently delivered us a new identity that it exerted anti-tumor activity through suppressing mTOR in various tumors. But the anti-tumor effect of metformin in AML was not clear. METHODS In this study, we used CCK8 assay and apoptosis assay to determine the anti-leukemia activity of metformin combined with AraC, and explore the mechanism of the joint role of Ara-C/metformin in AML. We finally used xenograft experiment in mice to determine the anti-leukemia effect of Ara-C/metformin in vivo. KEY FINDINGS We found that metformin could synergistically sensitize AML cells to Ara-C via inhibiting mTORC1/P70S6K pathway. In vivo experiment also verified metformin in aid of Ara-C caused an obviously synergistic anti-tumor effect. SIGNIFICANCE We firstly found the synergistic anti-tumor effect of Ara-C/metformin in AML through inhibiting mTORC1/P70S6K pathway.
Collapse
|
170
|
Ouabain impairs cancer metabolism and activates AMPK-Src signaling pathway in human cancer cell lines. Acta Pharmacol Sin 2020; 41:110-118. [PMID: 31515527 PMCID: PMC7468359 DOI: 10.1038/s41401-019-0290-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
In addition to the well-known cardiotonic effects, cardiac glycosides (CGs) produce potent anticancer effects with various molecular mechanisms. We previously show that ouabain induces autophagic cell death in human lung cancer cells by regulating AMPK-mediated mTOR and Src-mediated ERK1/2 signaling pathways. However, whether and how AMPK and Src signaling interacts in ouabain-treated cancer cells remains unclear. Given the pivotal role of AMPK in metabolism, whether ouabain affects cancer cell metabolism remains elusive. In this study we showed that treatment with ouabain (25 nM) caused simultaneous activation of AMPK and Src signaling pathways in human lung cancer A549 cells and human breast cancer MCF7 cells. Cotreatment with AMPK inhibitor compound C or siRNA greatly abrogates ouabain-induced Src activation, whereas cotreatment with Src inhibitor PP2 has little effect on ouabain-induced AMPK activity, suggesting that AMPK served as an upstream regulator of the Src signaling pathway. On the other hand, ouabain treatment greatly depletes ATP production in A549 and MCF7 cells, and supplement of ATP (100 μM) blocked ouabain-induced AMPK activation. We further demonstrated that ouabain greatly inhibited the mitochondrial oxidative phosphorylation (OXPHOS) in the cancer cells, and exerted differential metabolic effects on glycolysis depending on cancer cell type. Taken together, this study reveals that the altered cancer cell metabolism caused by ouabain may contribute to AMPK activation, as well as its cytotoxicity towards cancer cells.
Collapse
|
171
|
Wang Y, Nasiri AR, Damsky WE, Perry CJ, Zhang XM, Rabin-Court A, Pollak MN, Shulman GI, Perry RJ. Uncoupling Hepatic Oxidative Phosphorylation Reduces Tumor Growth in Two Murine Models of Colon Cancer. Cell Rep 2019; 24:47-55. [PMID: 29972790 DOI: 10.1016/j.celrep.2018.06.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/07/2018] [Accepted: 06/01/2018] [Indexed: 10/28/2022] Open
Abstract
Obesity is associated with colon cancer pathogenesis, but the underlying mechanism is actively debated. Here, we confirm that diet-induced obesity promotes tumor growth in two murine colon cancer models and show that this effect is reversed by an orally administered controlled-release mitochondrial protonophore (CRMP) that acts as a liver-specific uncoupler of oxidative phosphorylation. This agent lowered circulating insulin, and the reduction of tumor growth was abrogated by an insulin infusion raising plasma insulin to the level of high-fat-fed mice. We also demonstrate that hyperinsulinemia increases glucose uptake and oxidation in vivo in tumors and that CRMP reverses these effects. This study provides evidence that perturbations of whole-organism energy balance or hepatic energy metabolism can influence neoplastic growth. Furthermore, the data show that glucose uptake and utilization by cancers in vivo are not necessarily constitutively high but rather may vary according to the hormonal milieu.
Collapse
Affiliation(s)
- Yongliang Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ali R Nasiri
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - William E Damsky
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Curtis J Perry
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xian-Man Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Aviva Rabin-Court
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Michael N Pollak
- Department of Oncology, McGill University, Montreal, Quebec H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, Quebec H3T 1E2, Canada; Segal Cancer Centre, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
172
|
Luengo A, Abbott KL, Davidson SM, Hosios AM, Faubert B, Chan SH, Freinkman E, Zacharias LG, Mathews TP, Clish CB, DeBerardinis RJ, Lewis CA, Vander Heiden MG. Reactive metabolite production is a targetable liability of glycolytic metabolism in lung cancer. Nat Commun 2019; 10:5604. [PMID: 31811141 PMCID: PMC6898239 DOI: 10.1038/s41467-019-13419-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022] Open
Abstract
Increased glucose uptake and metabolism is a prominent phenotype of most cancers, but efforts to clinically target this metabolic alteration have been challenging. Here, we present evidence that lactoylglutathione (LGSH), a byproduct of methylglyoxal detoxification, is elevated in both human and murine non-small cell lung cancers (NSCLC). Methylglyoxal is a reactive metabolite byproduct of glycolysis that reacts non-enzymatically with nucleophiles in cells, including basic amino acids, and reduces cellular fitness. Detoxification of methylglyoxal requires reduced glutathione (GSH), which accumulates to high levels in NSCLC relative to normal lung. Ablation of the methylglyoxal detoxification enzyme glyoxalase I (Glo1) potentiates methylglyoxal sensitivity and reduces tumor growth in mice, arguing that targeting pathways involved in detoxification of reactive metabolites is an approach to exploit the consequences of increased glucose metabolism in cancer.
Collapse
Affiliation(s)
- Alba Luengo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Keene L Abbott
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shawn M Davidson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, 02142, USA
| | - Aaron M Hosios
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Brandon Faubert
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sze Ham Chan
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Elizaveta Freinkman
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Lauren G Zacharias
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas P Mathews
- 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
| | - Clary B Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA, 02142, USA
| | - 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
- Department of Pediatrics and Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Broad Institute of MIT and Harvard University, Cambridge, MA, 02142, USA.
- Dana-Farber Cancer Institute, Boston, MA, 02115, USA.
| |
Collapse
|
173
|
Buensuceso A, Ramos-Valdes Y, DiMattia GE, Shepherd TG. AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis. Mol Cancer Res 2019; 18:488-500. [PMID: 31744879 DOI: 10.1158/1541-7786.mcr-19-0530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/27/2019] [Accepted: 11/14/2019] [Indexed: 11/16/2022]
Abstract
Epithelial ovarian cancer (EOC) spreads by direct dissemination of malignant cells and multicellular clusters, known as spheroids, into the peritoneum followed by implantation and growth on abdominal surfaces. Using a spheroid model system of EOC metastasis, we discovered that Liver kinase B1 (LKB1), encoded by the STK11 gene, and its canonical substrate AMP-activated protein kinase (AMPK) are activated in EOC spheroids, yet only LKB1 is required for cell survival. We have now generated STK11-knockout cell lines using normal human FT190 cells and three EOC cell lines, OVCAR8, HeyA8, and iOvCa147. STK11KO did not affect growth and viability in adherent culture, but it decreased anchorage-independent growth of EOC cells. EOC spheroids lacking LKB1 had markedly impaired growth and viability, whereas there was no difference in normal FT190 spheroids. To test whether LKB1 loss affects EOC metastasis, we performed intraperitoneal injections of OVCAR8-, HeyA8-, and iOvCa147-STK11KO cells, and respective controls. LKB1 loss exhibited a dramatic reduction on tumor burden and metastatic potential; in particular, OVCAR8-STK11KO tumors had evidence of extensive necrosis, apoptosis, and hypoxia. Interestingly, LKB1 loss did not affect AMPKα phosphorylation in EOC spheroids and tumor xenografts, indicating that LKB1 signaling to support EOC cell survival in spheroids and metastatic tumor growth occurs via other downstream mediators. We identified the dual-specificity phosphatase DUSP4 as a commonly upregulated protein due to LKB1 loss; indeed, DUSP4 knockdown in HeyA8-STK11KO cells partially restored spheroid formation and viability. IMPLICATIONS: LKB1 possesses key tumor-promoting activity independent of downstream AMPK signaling during EOC metastasis.
Collapse
Affiliation(s)
- Adrian Buensuceso
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, Ontario, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Yudith Ramos-Valdes
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, Ontario, Canada
| | - Gabriel E DiMattia
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, Ontario, Canada.,Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Oncology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, Ontario, Canada. .,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Oncology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
174
|
Vanhove K, Graulus GJ, Mesotten L, Thomeer M, Derveaux E, Noben JP, Guedens W, Adriaensens P. The Metabolic Landscape of Lung Cancer: New Insights in a Disturbed Glucose Metabolism. Front Oncol 2019; 9:1215. [PMID: 31803611 PMCID: PMC6873590 DOI: 10.3389/fonc.2019.01215] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolism encompasses the biochemical processes that allow healthy cells to keep energy, redox balance and building blocks required for cell development, survival, and proliferation steady. Malignant cells are well-documented to reprogram their metabolism and energy production networks to support rapid proliferation and survival in harsh conditions via mutations in oncogenes and inactivation of tumor suppressor genes. Despite the histologic and genetic heterogeneity of tumors, a common set of metabolic pathways sustain the high proliferation rates observed in cancer cells. This review with a focus on lung cancer covers several fundamental principles of the disturbed glucose metabolism, such as the “Warburg” effect, the importance of the glycolysis and its branching pathways, the unanticipated gluconeogenesis and mitochondrial metabolism. Furthermore, we highlight our current understanding of the disturbed glucose metabolism and how this might result in the development of new treatments.
Collapse
Affiliation(s)
- Karolien Vanhove
- UHasselt, Faculty of Medicine and Life Sciences, LCRC, Diepenbeek, Belgium.,Department of Respiratory Medicine, Algemeen Ziekenhuis Vesalius, Tongeren, Belgium
| | - Geert-Jan Graulus
- Biomolecule Design Group, Institute for Materials Research, Hasselt University, Diepenbeek, Belgium
| | - Liesbet Mesotten
- UHasselt, Faculty of Medicine and Life Sciences, LCRC, Diepenbeek, Belgium.,Department of Nuclear Medicine, Ziekenhuis Oost Limburg, Genk, Belgium
| | - Michiel Thomeer
- UHasselt, Faculty of Medicine and Life Sciences, LCRC, Diepenbeek, Belgium.,Department of Respiratory Medicine, Ziekenhuis Oost Limburg, Genk, Belgium
| | - Elien Derveaux
- UHasselt, Faculty of Medicine and Life Sciences, LCRC, Diepenbeek, Belgium
| | - Jean-Paul Noben
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research, Hasselt University, Diepenbeek, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research, Hasselt University, Diepenbeek, Belgium.,Applied and Analytical Chemistry, Institute for Materials Research, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
175
|
Chen F, Alphonse MP, Liu Y, Liu Q. Targeting Mutant KRAS for Anticancer Therapy. Curr Top Med Chem 2019; 19:2098-2113. [DOI: 10.2174/1568026619666190902151307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
:Over the past decades, designing therapeutic strategies to target KRAS-mutant cancers, which is one of the most frequent mutant oncogenes among all cancer types, have proven unsuccessful regardless of many concerted attempts. There are key challenges for KRAS-mutant anticancer therapy, as the complex cellular processes involved in KRAS signaling has present. Herein, we highlight the emerging therapeutic approaches for inhibiting KRAS signaling and blocking KRAS functions, in hope to serve as a more effective guideline for future development of therapeutics.
Collapse
Affiliation(s)
- Fengqian Chen
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, TX 79416, United States
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 216, 1550 Orleans Street, Baltimore, MD 21231, United States
| | - Yan Liu
- Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766, United States
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 216, 1550 Orleans Street, Baltimore, MD 21231, United States
| |
Collapse
|
176
|
The anti-cancer effects of phenformin in thyroid cancer cell lines and in normal thyrocytes. Oncotarget 2019; 10:6432-6443. [PMID: 31741708 PMCID: PMC6849649 DOI: 10.18632/oncotarget.27266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/24/2019] [Indexed: 12/26/2022] Open
Abstract
Phenformin is a biguanide drug which, besides the original anti-diabetic effect, also exerts anti-cancer effects. The aim of this study was to further characterize these latter in terms of both cell-viability and modulation of the secretion of the pro-tumorigenic chemokine CXCL8. Normal human thyrocytes in primary cultures (NHT) and thyroid cancer cell lines, TPC-1 and 8505C (RET/PTC and BRAFV600E mutated, respectively) were treated with increasing concentrations of phenformin at different times. Cell-viability was assessed by WST-1 and further characterized by AnnexinV/PI staining and cell proliferation colony-assay. CXCL8 levels were measured in cell supernatants. Phenformin reduced cell-viability in TPC-1 and 8505C and their ability to form colonies. In NHT cells, phenformin affected cell-viability only at the maximal dose but interestingly it inhibited CXCL8 secretion at all the concentrations not affecting cell-viability. Phenformin had no effect on CXCL8 secretion in thyroid cancer cell lines. Thus, phenformin exerts anti-cancer effects on both cancer cells (cell death induction) and surrounding normal cells (inhibition of CXCL8 secretion). These results highlight that the anti-cancer effects of phenformin are multifaceted and effective on both solid and soluble components of the tumor-microenvironment.
Collapse
|
177
|
LKB1/AMPK Pathway and Drug Response in Cancer: A Therapeutic Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8730816. [PMID: 31781355 PMCID: PMC6874879 DOI: 10.1155/2019/8730816] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 12/25/2022]
Abstract
Inactivating mutations of the tumor suppressor gene Liver Kinase B1 (LKB1) are frequently detected in non-small-cell lung cancer (NSCLC) and cervical carcinoma. Moreover, LKB1 expression is epigenetically regulated in several tumor types. LKB1 has an established function in the control of cell metabolism and oxidative stress. Clinical and preclinical studies support a role of LKB1 as a central modifier of cellular response to different stress-inducing drugs, suggesting LKB1 pathway as a highly promising therapeutic target. Loss of LKB1-AMPK signaling confers sensitivity to energy depletion and to redox homeostasis impairment and has been associated with an improved outcome in advanced NSCLC patients treated with chemotherapy. In this review, we provide an overview of the interplay between LKB1 and its downstream targets in cancer and focus on potential therapeutic strategies whose outcome could depend from LKB1.
Collapse
|
178
|
Momcilovic M, Jones A, Bailey ST, Waldmann CM, Li R, Lee JT, Abdelhady G, Gomez A, Holloway T, Schmid E, Stout D, Fishbein MC, Stiles L, Dabir DV, Dubinett SM, Christofk H, Shirihai O, Koehler CM, Sadeghi S, Shackelford DB. In vivo imaging of mitochondrial membrane potential in non-small-cell lung cancer. Nature 2019; 575:380-384. [PMID: 31666695 DOI: 10.1038/s41586-019-1715-0] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 09/26/2019] [Indexed: 12/21/2022]
Abstract
Mitochondria are essential regulators of cellular energy and metabolism, and have a crucial role in sustaining the growth and survival of cancer cells. A central function of mitochondria is the synthesis of ATP by oxidative phosphorylation, known as mitochondrial bioenergetics. Mitochondria maintain oxidative phosphorylation by creating a membrane potential gradient that is generated by the electron transport chain to drive the synthesis of ATP1. Mitochondria are essential for tumour initiation and maintaining tumour cell growth in cell culture and xenografts2,3. However, our understanding of oxidative mitochondrial metabolism in cancer is limited because most studies have been performed in vitro in cell culture models. This highlights a need for in vivo studies to better understand how oxidative metabolism supports tumour growth. Here we measure mitochondrial membrane potential in non-small-cell lung cancer in vivo using a voltage-sensitive, positron emission tomography (PET) radiotracer known as 4-[18F]fluorobenzyl-triphenylphosphonium (18F-BnTP)4. By using PET imaging of 18F-BnTP, we profile mitochondrial membrane potential in autochthonous mouse models of lung cancer, and find distinct functional mitochondrial heterogeneity within subtypes of lung tumours. The use of 18F-BnTP PET imaging enabled us to functionally profile mitochondrial membrane potential in live tumours.
Collapse
Affiliation(s)
- Milica Momcilovic
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Anthony Jones
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Sean T Bailey
- The Mouse Phase I Unit, Lineberger School of Medicine at the University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Christopher M Waldmann
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Rui Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Jason T Lee
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Crump Institute for Molecular Imaging, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Gihad Abdelhady
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Adrian Gomez
- Department of Chemistry and Biochemistry, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Travis Holloway
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Ernst Schmid
- Department of Biological Chemistry, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | | | - Michael C Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Endocrinology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Deepa V Dabir
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Heather Christofk
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Department of Biological Chemistry, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,UCLA Metabolomics Center, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Orian Shirihai
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.,Department of Endocrinology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Carla M Koehler
- Department of Chemistry and Biochemistry, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Saman Sadeghi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - David B Shackelford
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.
| |
Collapse
|
179
|
Quintela-Fandino M, Morales S, Cortés-Salgado A, Manso L, Apala JV, Muñoz M, Gasol Cudos A, Salla Fortuny J, Gion M, Lopez-Alonso A, Cortés J, Guerra J, Malón D, Caleiras E, Mulero F, Mouron S. Randomized Phase 0/I Trial of the Mitochondrial Inhibitor ME-344 or Placebo Added to Bevacizumab in Early HER2-Negative Breast Cancer. Clin Cancer Res 2019; 26:35-45. [DOI: 10.1158/1078-0432.ccr-19-2023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/09/2019] [Accepted: 10/03/2019] [Indexed: 11/16/2022]
|
180
|
Park J, Shim JK, Kang JH, Choi J, Chang JH, Kim SY, Kang SG. Regulation of bioenergetics through dual inhibition of aldehyde dehydrogenase and mitochondrial complex I suppresses glioblastoma tumorspheres. Neuro Oncol 2019; 20:954-965. [PMID: 29294080 DOI: 10.1093/neuonc/nox243] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Targeted approaches for treating glioblastoma (GBM) attempted to date have consistently failed, highlighting the imperative for treatment strategies that operate on different mechanistic principles. Bioenergetics deprivation has emerged as an effective therapeutic approach for various tumors. We have previously found that cancer cells preferentially utilize cytosolic NADH supplied by aldehyde dehydrogenase (ALDH) for ATP production through oxidative phosphorylation (OxPhos). This study is aimed at examining therapeutic responses and underlying mechanisms of dual inhibition of ALDH and OxPhos against GBM. Methods For inhibition of ALDH and OxPhos, the corresponding inhibitors, gossypol and phenformin were used. Biological functions, including ATP levels, stemness, invasiveness, and viability, were evaluated in GBM tumorspheres (TSs). Gene expression profiles were analyzed using microarray data. In vivo anticancer efficacy was examined in a mouse orthotopic xenograft model. Results Combined treatment of GBM TSs with gossypol and phenformin significantly reduced ATP levels, stemness, invasiveness, and cell viability. Consistently, this therapy substantially decreased expression of genes associated with stemness, mesenchymal transition, and invasion in GBM TSs. Supplementation of ATP using malate abrogated these effects, whereas knockdown of ALDH1L1 mimicked them, suggesting that disruption of ALDH-mediated ATP production is a key mechanism of this therapeutic combination. In vivo efficacy confirmed remarkable therapeutic responses to combined treatment with gossypol and phenformin. Conclusion Our findings suggest that dual inhibition of tumor bioenergetics is a novel and effective strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Junseong Park
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Kyoung Shim
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Hee Kang
- Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo-Youl Kim
- Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
181
|
Bokil A, Sancho P. Mitochondrial determinants of chemoresistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:634-646. [PMID: 35582564 PMCID: PMC8992520 DOI: 10.20517/cdr.2019.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022]
Abstract
Chemoresistance constitute nowadays the major contributor to therapy failure in most cancers. There are main factors that mitigate cell response to therapy, such as target organ, inherent sensitivity to the administered compound, its metabolism, drug efflux and influx or alterations on specific cellular targets, among others. We now know that intrinsic properties of cancer cells, including metabolic features, substantially contribute to chemoresistance. In fact, during the last years, numerous reports indicate that cancer cells resistant to chemotherapy demonstrate significant alterations in mitochondrial metabolism, membrane polarization and mass. Metabolic activity and expression of several mitochondrial proteins are modulated under treatment to cope with stress, making these organelles central players in the development of resistance to therapies. Here, we review the role of mitochondria in chemoresistant cells in terms of metabolic rewiring and function of key mitochondria-related proteins.
Collapse
Affiliation(s)
- Ansooya Bokil
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| | - Patricia Sancho
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| |
Collapse
|
182
|
Abstract
Tumors display reprogrammed metabolic activities that promote cancer progression. We currently possess a limited understanding of the processes governing tumor metabolism in vivo and of the most efficient approaches to identify metabolic vulnerabilities susceptible to therapeutic targeting. While much of the literature focuses on stereotyped, cell-autonomous pathways like glycolysis, recent work emphasizes heterogeneity and flexibility of metabolism between tumors and even within distinct regions of solid tumors. Metabolic heterogeneity is important because it influences therapeutic vulnerabilities and may predict clinical outcomes. This Review describes current concepts about metabolic regulation in tumors, focusing on processes intrinsic to cancer cells and on factors imposed upon cancer cells by the tumor microenvironment. We discuss experimental approaches to identify subtype-selective metabolic vulnerabilities in preclinical cancer models. Finally, we describe efforts to characterize metabolism in primary human tumors, which should produce new insights into metabolic heterogeneity in the context of clinically relevant microenvironments.
Collapse
Affiliation(s)
- Jiyeon Kim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
183
|
Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer 2019; 19:495-509. [PMID: 31406302 PMCID: PMC7043073 DOI: 10.1038/s41568-019-0179-8] [Citation(s) in RCA: 654] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 02/07/2023]
Abstract
The impressive clinical activity of small-molecule receptor tyrosine kinase inhibitors for oncogene-addicted subgroups of non-small-cell lung cancer (for example, those driven by activating mutations in the gene encoding epidermal growth factor receptor (EGFR) or rearrangements in the genes encoding the receptor tyrosine kinases anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 (ROS1) and rearranged during transfection (RET)) has established an oncogene-centric molecular classification paradigm in this disease. However, recent studies have revealed considerable phenotypic diversity downstream of tumour-initiating oncogenes. Co-occurring genomic alterations, particularly in tumour suppressor genes such as TP53 and LKB1 (also known as STK11), have emerged as core determinants of the molecular and clinical heterogeneity of oncogene-driven lung cancer subgroups through their effects on both tumour cell-intrinsic and non-cell-autonomous cancer hallmarks. In this Review, we discuss the impact of co-mutations on the pathogenesis, biology, microenvironmental interactions and therapeutic vulnerabilities of non-small-cell lung cancer and assess the challenges and opportunities that co-mutations present for personalized anticancer therapy, as well as the expanding field of precision immunotherapy.
Collapse
Affiliation(s)
- Ferdinandos Skoulidis
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
184
|
Bhatia S, Monkman J, Blick T, Duijf PH, Nagaraj SH, Thompson EW. Multi-Omics Characterization of the Spontaneous Mesenchymal-Epithelial Transition in the PMC42 Breast Cancer Cell Lines. J Clin Med 2019; 8:E1253. [PMID: 31430931 PMCID: PMC6723942 DOI: 10.3390/jcm8081253] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 12/16/2022] Open
Abstract
Epithelial-mesenchymal plasticity (EMP), encompassing epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET), are considered critical events for cancer metastasis. We investigated chromosomal heterogeneity and chromosomal instability (CIN) profiles of two sister PMC42 breast cancer (BC) cell lines to assess the relationship between their karyotypes and EMP phenotypic plasticity. Karyotyping by GTG banding and exome sequencing were aligned with SWATH quantitative proteomics and existing RNA-sequencing data from the two PMC42 cell lines; the mesenchymal, parental PMC42-ET cell line and the spontaneously epithelially shifted PMC42-LA daughter cell line. These morphologically distinct PMC42 cell lines were also compared with five other BC cell lines (MDA-MB-231, SUM-159, T47D, MCF-7 and MDA-MB-468) for their expression of EMP and cell surface markers, and stemness and metabolic profiles. The findings suggest that the epithelially shifted cell line has a significantly altered ploidy of chromosomes 3 and 13, which is reflected in their transcriptomic and proteomic expression profiles. Loss of the TGFβR2 gene from chromosome 3 in the epithelial daughter cell line inhibits its EMT induction by TGF-β stimulus. Thus, integrative 'omics' characterization established that the PMC42 system is a relevant MET model and provides insights into the regulation of phenotypic plasticity in breast cancer.
Collapse
Affiliation(s)
- Sugandha Bhatia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.
- Translational Research Institute, Brisbane, QLD 4102, Australia.
| | - James Monkman
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Tony Blick
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Pascal Hg Duijf
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Shivashankar H Nagaraj
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.
- Translational Research Institute, Brisbane, QLD 4102, Australia.
| |
Collapse
|
185
|
Park JH, Kim YH, Park EH, Lee SJ, Kim H, Kim A, Lee SB, Shim S, Jang H, Myung JK, Park S, Lee SJ, Kim MJ. Effects of metformin and phenformin on apoptosis and epithelial-mesenchymal transition in chemoresistant rectal cancer. Cancer Sci 2019; 110:2834-2845. [PMID: 31278880 PMCID: PMC6726705 DOI: 10.1111/cas.14124] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022] Open
Abstract
Recurrence and chemoresistance in colorectal cancer remain important issues for patients treated with conventional therapeutics. Metformin and phenformin, previously used in the treatment of diabetes, have been shown to have anticancer effects in various cancers, including breast, lung and prostate cancers. However, their molecular mechanisms are still unclear. In this study, we examined the effects of these drugs in chemoresistant rectal cancer cell lines. We found that SW837 and SW1463 rectal cancer cells were more resistant to ionizing radiation and 5‐fluorouracil than HCT116 and LS513 colon cancer cells. In addition, metformin and phenformin increased the sensitivity of these cell lines by inhibiting cell proliferation, suppressing clonogenic ability and increasing apoptotic cell death in rectal cancer cells. Signal transducer and activator of transcription 3 and transforming growth factor‐β/Smad signaling pathways were more activated in rectal cancer cells, and inhibition of signal transducer and activator of transcription 3 expression using an inhibitor or siRNA sensitized rectal cancer cells to chemoresistant by inhibition of the expression of antiapoptotic proteins, such as X‐linked inhibitor of apoptosis, survivin and cellular inhibitor of apoptosis protein 1. Moreover, metformin and phenformin inhibited cell migration and invasion by suppression of transforming growth factor β receptor 2‐mediated Snail and Twist expression in rectal cancer cells. Therefore, metformin and phenformin may represent a novel strategy for the treatment of chemoresistant rectal cancer by targeting signal transducer and activator of transcription 3 and transforming growth factor‐β/Smad signaling.
Collapse
Affiliation(s)
- Ji-Hye Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Young-Heon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Eun Hyeh Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sun-Joo Lee
- Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Hyewon Kim
- Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Areumnuri Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea.,Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea.,Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Min Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| |
Collapse
|
186
|
Zhang K, Wang J, Wang J, Luh F, Liu X, Yang L, Liu YR, Su L, Yang YCSH, Chu P, Yen Y. LKB1 deficiency promotes proliferation and invasion of glioblastoma through activation of mTOR and focal adhesion kinase signaling pathways. Am J Cancer Res 2019; 9:1650-1663. [PMID: 31497348 PMCID: PMC6726989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/28/2019] [Indexed: 06/10/2023] Open
Abstract
Liver kinase B1 (LKB1), a serine/threonine kinase, is frequently inactivated in several types of human cancers. To date, inactivation of LKB1 tumor suppressor has rarely been reported in glioblastoma. In this study, we investigated LKB1 status, biological significance, and therapeutic implications in glioblastoma. Loss of LKB1 immunostaining was identified in 8.6% (5/58), while decrease of LKB1 immunostaining was found in 29.3% (17/58) of glioblastoma tissues. Notably, mining TCGA database of LKB1 expression in glioblastoma revealed that lower mRNA level of LKB1 was associated with shorter survival in glioblastoma. We found that knockdown of LKB1 significantly promoted in vitro proliferation, adhesion, invasion, and metformin-induced apoptosis, and simultaneously enhanced activation of ERK and mammalian-target of rapamycin (mTOR) signaling pathways in LKB1-compenent U87 and T98 glioblastoma cells. Moreover, global transcriptional profiling revealed that adhesion and cytoskeletal proteins such as Vinculin, Talin and signaling pathways including focal adhesion kinase (FAK), extracellular martrix (ECM) receptor interaction, and cellular motility were significantly enriched in U87 and T98 glioblastoma cells upon LKB1 knockdown. Additionally, we demonstrated that the enhanced activation of FAK by LKB1 knockdown was dependent on differentially expressed cytoskeletal proteins in these glioblastoma cells. Importantly, we further found that mTOR1 inhibitor rapamycin dominantly inhibited in vitro cellular proliferation, while FAK inhibitor PF-573288 drastically decreased invasion of LKB1-attenuated glioblastoma cells. Therefore, downregulation of LKB1 may contribute to the pathogenesis and malignancy of glioblastoma and may have potential implications for stratification and treatment of glioblastoma patients.
Collapse
Affiliation(s)
- Keqiang Zhang
- Department of Surgery, City of Hope National Medical CenterDuarte, CA, USA
| | - Jinghan Wang
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical HospitalShanghai, China
| | - Jinhui Wang
- The Integrative Genomics Core Lab, City of Hope National Medical CenterDuarte, CA, USA
| | - Frank Luh
- Sino-American Cancer FoundationTemple City, CA, USA
| | - Xiyong Liu
- Sino-American Cancer FoundationTemple City, CA, USA
| | - Lu Yang
- Department of System Biology, City of Hope National Medical CenterDuarte, CA, USA
| | - Yun-Ru Liu
- Comprehensive Cancer Center, Taipei Medical UniversityTaipei, Taiwan
| | - Leila Su
- Sino-American Cancer FoundationTemple City, CA, USA
| | - Yu-Chen SH Yang
- PhD Program of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical UniversityTaipei, Taiwan
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical CenterDuarte, CA, USA
| | - Yun Yen
- Comprehensive Cancer Center, Taipei Medical UniversityTaipei, Taiwan
- PhD Program of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical UniversityTaipei, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical UniversityTaipei, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical UniversityTaipei, Taiwan
| |
Collapse
|
187
|
Hollstein PE, Eichner LJ, Brun SN, Kamireddy A, Svensson RU, Vera LI, Ross DS, Rymoff TJ, Hutchins A, Galvez HM, Williams AE, Shokhirev MN, Screaton RA, Berdeaux R, Shaw RJ. The AMPK-Related Kinases SIK1 and SIK3 Mediate Key Tumor-Suppressive Effects of LKB1 in NSCLC. Cancer Discov 2019; 9:1606-1627. [PMID: 31350328 DOI: 10.1158/2159-8290.cd-18-1261] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 05/29/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
Mutations in the LKB1 (also known as STK11) tumor suppressor are the third most frequent genetic alteration in non-small cell lung cancer (NSCLC). LKB1 encodes a serine/threonine kinase that directly phosphorylates and activates 14 AMPK family kinases ("AMPKRs"). The function of many of the AMPKRs remains obscure, and which are most critical to the tumor-suppressive function of LKB1 remains unknown. Here, we combine CRISPR and genetic analysis of the AMPKR family in NSCLC cell lines and mouse models, revealing a surprising critical role for the SIK subfamily. Conditional genetic loss of Sik1 revealed increased tumor growth in mouse models of Kras-dependent lung cancer, which was further enhanced by loss of the related kinase Sik3. As most known substrates of the SIKs control transcription, gene-expression analysis was performed, revealing upregulation of AP1 and IL6 signaling in common between LKB1- and SIK1/3-deficient tumors. The SIK substrate CRTC2 was required for this effect, as well as for proliferation benefits from SIK loss. SIGNIFICANCE: The tumor suppressor LKB1/STK11 encodes a serine/threonine kinase frequently inactivated in NSCLC. LKB1 activates 14 downstream kinases in the AMPK family controlling growth and metabolism, although which kinases are critical for LKB1 tumor-suppressor function has remained an enigma. Here we unexpectedly found that two understudied kinases, SIK1 and SIK3, are critical targets in lung cancer.This article is highlighted in the In This Issue feature, p. 1469.
Collapse
Affiliation(s)
- Pablo E Hollstein
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Lillian J Eichner
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Sonja N Brun
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Anwesh Kamireddy
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Robert U Svensson
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Liliana I Vera
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Debbie S Ross
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - T J Rymoff
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Amanda Hutchins
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - Hector M Galvez
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California
| | - April E Williams
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, California
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, California
| | - Robert A Screaton
- Sunnybrook Research Institute and Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Reuben J Shaw
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California.
| |
Collapse
|
188
|
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: 105] [Impact Index Per Article: 17.5] [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
|
189
|
Visnjic D, Dembitz V, Lalic H. The Role of AMPK/mTOR Modulators in the Therapy of Acute Myeloid Leukemia. Curr Med Chem 2019; 26:2208-2229. [PMID: 29345570 DOI: 10.2174/0929867325666180117105522] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 01/01/2018] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Differentiation therapy of acute promyelocytic leukemia with all-trans retinoic acid represents the most successful pharmacological therapy of acute myeloid leukemia (AML). Numerous studies demonstrate that drugs that inhibit mechanistic target of rapamycin (mTOR) and activate AMP-kinase (AMPK) have beneficial effects in promoting differentiation and blocking proliferation of AML. Most of these drugs are already in use for other purposes; rapalogs as immunosuppressants, biguanides as oral antidiabetics, and 5-amino-4-imidazolecarboxamide ribonucleoside (AICAr, acadesine) as an exercise mimetic. Although most of these pharmacological modulators have been widely used for decades, their mechanism of action is only partially understood. In this review, we summarize the role of AMPK and mTOR in hematological malignancies and discuss the possible role of pharmacological modulators in proliferation and differentiation of leukemia cells.
Collapse
Affiliation(s)
- Dora Visnjic
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 12, 10 000 Zagreb, Croatia
| | - Vilma Dembitz
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 12, 10 000 Zagreb, Croatia
| | - Hrvoje Lalic
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 12, 10 000 Zagreb, Croatia
| |
Collapse
|
190
|
Heinz S, Freyberger A, Lawrenz B, Schladt L, Schmuck G, Ellinger-Ziegelbauer H. Energy metabolism modulation by biguanides in comparison with rotenone in rat liver and heart. Arch Toxicol 2019; 93:2603-2615. [DOI: 10.1007/s00204-019-02519-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022]
|
191
|
Bie F, Wang G, Qu X, Wang Y, Huang C, Wang Y, Du J. Loss of FGL1 induces epithelial‑mesenchymal transition and angiogenesis in LKB1 mutant lung adenocarcinoma. Int J Oncol 2019; 55:697-707. [PMID: 31322182 DOI: 10.3892/ijo.2019.4838] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 07/04/2019] [Indexed: 12/31/2022] Open
Abstract
Liver kinase b1 (LKB1) is a tumor suppressor, and the inactivated mutation frequency of LKB1 in lung adenocarcinoma is ~20%. The present study aimed to explore potential novel biomarkers in LKB1 mutant lung adenocarcinoma. Gene expression data from lung adenocarcinoma patients were downloaded from The Cancer Genome Atlas and the Gene Expression Omnibus databases. R software was used to analyze the gene expression profiles. Reverse transcription‑quantitative PCR (RT‑qPCR), western blot and immunohistochemistry (IHC) analyses were used to examine gene expression and function. Gene function was further explored via gene set enrichment analysis. A colony formation assay was used to evaluate cell proliferation. A wound‑healing assay and immunofluorescence analysis were used to evaluate cell migration and epithelial‑mesenchymal transition (EMT), respectively. Wound healing assay, immunofluorescence, western blot, RT‑qPCR and IHC results for EMT‑associated markers demonstrated that a loss of fibrinogen‑like 1 (FGL1) induced EMT in LKB1 mutant lung adenocarcinoma. RT‑qPCR and IHC analyses of angiogenesis‑related markers revealed that loss of FGL1 promoted angiogenesis in LKB1 mutant lung adenocarcinoma. Overall, the present results demonstrated that loss of FGL1 induced EMT and angiogenesis in LKB1 mutant lung adenocarcinoma. FGL1 may be a novel biomarker to indicate EMT and angiogenesis in patients with LKB1 mutant lung adenocarcinoma.
Collapse
Affiliation(s)
- Fenglong Bie
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Guanghui Wang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiao Qu
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yadong Wang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Cuicui Huang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yu Wang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
192
|
Phenformin as an Anticancer Agent: Challenges and Prospects. Int J Mol Sci 2019; 20:ijms20133316. [PMID: 31284513 PMCID: PMC6651400 DOI: 10.3390/ijms20133316] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022] Open
Abstract
Currently, there is increasing evidence linking diabetes mellitus (especially type 2 diabetes mellitus) with carcinogenesis through various biological processes, such as fat-induced chronic inflammation, hyperglycemia, hyperinsulinemia, and angiogenesis. Chemotherapeutic agents are used in the treatment of cancer, but in most cases, patients develop resistance. Phenformin, an oral biguanide drug used to treat type 2 diabetes mellitus, was removed from the market due to a high risk of fatal lactic acidosis. However, it has been shown that phenformin is, with other biguanides, an authentic tumor disruptor, not only by the production of hypoglycemia due to caloric restriction through AMP-activated protein kinase with energy detection (AMPK) but also as a blocker of the mTOR regulatory complex. Moreover, the addition of phenformin eliminates resistance to antiangiogenic tyrosine kinase inhibitors (TKI), which prevent the uncontrolled metabolism of glucose in tumor cells. In this review, we evidence the great potential of phenformin as an anticancer agent. We thoroughly review its mechanism of action and clinical trial assays, specially focusing on current challenges and future perspectives of this promising drug.
Collapse
|
193
|
Majem B, Nadal E, Muñoz-Pinedo C. Exploiting metabolic vulnerabilities of Non small cell lung carcinoma. Semin Cell Dev Biol 2019; 98:54-62. [PMID: 31238096 DOI: 10.1016/j.semcdb.2019.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022]
Abstract
Lung cancer is the main cause of cancer death worldwide. Non-Small Cell Lung Carcinoma (NSCLC) is the most common subtype of lung cancer, and the prognosis of NSCLC patients in advanced stages is still very poor. Given the need for new therapies, the metabolism of NSCLC has been widely studied in the past two decades to identify vulnerabilities that could be translated into novel anti-metabolic therapeutic approaches. A number of studies have highlighted the role of glucose and mitochondrial metabolism in the development of NSCLC. The metabolic properties of lung tumors have been characterized in detail in vivo, and they include high glucose and lactate use and high heterogeneity regarding the use of nutrients and mitochondrial pathways. This heterogeneity has also been observed in patients infused with labeled nutrients. We will summarize here the knowledge about the use of amino acids, fatty acids and carbohydrates in NSCLC that could lead to new combination treatments.
Collapse
Affiliation(s)
- Blanca Majem
- Cell Death Regulation Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Spain.
| | - Ernest Nadal
- Clinical Research in Solid Tumors (CReST) Group, Oncobell Program, IDIBELL, L'Hospitalet, Spain; Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet, Spain.
| | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Spain.
| |
Collapse
|
194
|
Rodón L, Svensson RU, Wiater E, Chun MGH, Tsai WW, Eichner LJ, Shaw RJ, Montminy M. The CREB coactivator CRTC2 promotes oncogenesis in LKB1-mutant non-small cell lung cancer. SCIENCE ADVANCES 2019; 5:eaaw6455. [PMID: 31355336 PMCID: PMC6656544 DOI: 10.1126/sciadv.aaw6455] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 06/17/2019] [Indexed: 05/15/2023]
Abstract
The LKB1 tumor suppressor is often mutationally inactivated in non-small cell lung cancer (NSCLC). LKB1 phosphorylates and activates members of the AMPK family of Ser/Thr kinases. Within this family, the salt-inducible kinases (SIKs) modulate gene expression in part via the inhibitory phosphorylation of the CRTCs, coactivators for CREB (cAMP response element-binding protein). The loss of LKB1 causes SIK inactivation and the induction of the CRTCs, leading to the up-regulation of CREB target genes. We identified CRTC2 as a critical factor in LKB1-deficient NSCLC. CRTC2 is unphosphorylated and therefore constitutively activated in LKB1-mutant NSCLC, where it promotes tumor growth, in part via the induction of the inhibitor of DNA binding 1 (ID1), a bona fide CREB target gene. As ID1 expression is up-regulated and confers poor prognosis in LKB1-deficient NSCLC, our results suggest that small molecules that inhibit CRTC2 and ID1 activity may provide therapeutic benefit to individuals with NSCLC.
Collapse
Affiliation(s)
- Laura Rodón
- Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Robert U. Svensson
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037 USA
| | - Ezra Wiater
- Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Matthew G. H Chun
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037 USA
| | - Wen-Wei Tsai
- Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Lillian J. Eichner
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037 USA
| | - Reuben J. Shaw
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037 USA
| | - Marc Montminy
- Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Corresponding author.
| |
Collapse
|
195
|
Ren Y, Shen HM. Critical role of AMPK in redox regulation under glucose starvation. Redox Biol 2019; 25:101154. [PMID: 30853530 PMCID: PMC6859544 DOI: 10.1016/j.redox.2019.101154] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/09/2019] [Accepted: 02/27/2019] [Indexed: 12/24/2022] Open
Abstract
Glucose starvation is one of the major forms of metabolic stress in cancer cells. Deprivation of glucose impairs glycolysis and the pentose phosphate pathway, which elicits oxidative stress due to enhanced production of reactive oxygen species (ROS) and impaired antioxidant system, leading to redox imbalance and cell death. Under glucose starvation, the 5' AMP-activated protein kinase (AMPK) plays a critical role in maintaining redox homeostasis and cell survival via multiple pathways, such as regulation of fatty acid metabolism and antioxidant response. Convergence of ROS and the glucose metabolic pathway reveals novel molecular targets for the development of effective cancer therapeutic strategies. Interestingly, AMPK, along with its upstream kinase liver kinase B1 (LKB1), has been regarded to play a tumor suppressor role. However, emerging studies have provided novel insights into the pro-tumor survival function of the LKB1-AMPK pathway. Therefore, targeting metabolic and oxidative stress in cancer cells, with manipulation of AMPK activity, is a promising strategy in developing novel cancer therapeutic agents.
Collapse
Affiliation(s)
- Yi Ren
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore.
| |
Collapse
|
196
|
Lee M, Hirpara JL, Eu JQ, Sethi G, Wang L, Goh BC, Wong AL. Targeting STAT3 and oxidative phosphorylation in oncogene-addicted tumors. Redox Biol 2019; 25:101073. [PMID: 30594485 PMCID: PMC6859582 DOI: 10.1016/j.redox.2018.101073] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/08/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
Drug resistance invariably limits the response of oncogene-addicted cancer cells to targeted therapy. The upregulation of signal transducer and activator of transcription 3 (STAT3) has been implicated as a mechanism of drug resistance in a range of oncogene-addicted cancers. However, the development of inhibitors against STAT3 has been fraught with challenges such as poor delivery or lack of specificity. Clinical experience with small molecule STAT3 inhibitors has seen efficacy signals, but this success has been tempered by drug limiting toxicities from off-target adverse events. It has emerged in recent years that, contrary to the Warburg theory, certain tumor types undergo metabolic reprogramming towards oxidative phosphorylation (OXPHOS) to satisfy their energy production. In particular, certain drug-resistant oncogene-addicted tumors have been found to rely on OXPHOS as a mechanism of survival. Multiple cellular signaling pathways converge on STAT3, hence the localization of STAT3 to the mitochondria may provide the link between oncogene-induced signaling pathways and cancer cell metabolism. In this article, we review the role of STAT3 and OXPHOS as targets of novel therapeutic strategies aimed at restoring drug sensitivity in treatment-resistant oncogene-addicted tumor types. Apart from drugs which have been re-purposed as OXPHOS inhibitors for-anti-cancer therapy (e.g., metformin and phenformin), several novel compounds in the drug-development pipeline have demonstrated promising pre-clinical and clinical activity. However, the clinical development of OXPHOS inhibitors remains in its infancy. The further identification of compounds with acceptable toxicity profiles, alongside the discovery of robust companion biomarkers of OXPHOS inhibition, would represent tangible early steps in transforming the therapeutic landscape of cancer cell metabolism.
Collapse
Affiliation(s)
- Matilda Lee
- Department of Haematology-Oncology, National University Health System, Singapore; Haematology-Oncology Research Group, National University Cancer Institute of Singapore, National University Health System, Singapore
| | | | | | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Boon-Cher Goh
- Department of Haematology-Oncology, National University Health System, Singapore; Haematology-Oncology Research Group, National University Cancer Institute of Singapore, National University Health System, Singapore; Cancer Science Institute, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Andrea L Wong
- Department of Haematology-Oncology, National University Health System, Singapore; Haematology-Oncology Research Group, National University Cancer Institute of Singapore, National University Health System, Singapore; Cancer Science Institute, Singapore.
| |
Collapse
|
197
|
Zarredar H, Farajnia S, Ansarin K, Baradaran B, Aria M, Asadi M. Synergistic Effect of Novel EGFR Inhibitor AZD8931 and p38α siRNA in Lung Adenocarcinoma Cancer Cells. Anticancer Agents Med Chem 2019; 19:638-644. [DOI: 10.2174/1871520619666190301125203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/26/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
Background:Lung cancer is the leading cause of cancer-related death with less than 5-year survival rate for both men and women worldwide. EGFR and MAPK signaling pathways have a critical role in proliferation and progression of various cancers, including lung cancer. P38 map kinase plays different role in various tissue hence showing a tissue-dependent behavior. It acts as an oncogene in some tissues while plays as tumor suppressor in some other tissues. The aim of this study was to investigate the combined effect of P38 αspecific siRNA and EGFR inhibitor on apoptosis and proliferation of A549 lung cancer cell line.Objective:This article is dedicated to the synergistic effect of novel EGFR inhibitor AZD8931 and P38 α siRNA in lung adenocarcinoma cancer cells proliferation and apoptosis.Methods and Materials:The A549 lung cancer cells were treated with P38 α- siRNA and EGFR inhibitor alone or in combination. The cytotoxic effects of P38 α- siRNA and EGFR inhibitor were determined using MTT assay. Relative P38 α and EGFR mRNA levels were measured by QRT-PCR. Induction of apoptosis were measured by FACS analysis.Results:The expression of mRNA related to P38 α, EGFR, and Her2 genes was reduced to 23.4%, 52.4%, and 75, respectively, after treatment of their inhibitors. Also, MTT assay showed that the cell viability after treatment with p38 α SiRNA, EGFR inhibitor and their combination was reduced to 51.02%, 48.9%, and 25.11%, respectively. FACS results indicated that p38 α siRNA, EGFR inhibitor and their combination, reduced the population of live cells to 49.5%, 32.2% and 14.3% in comparison to the population of untreated control cells (99.5%).Conclusion:The results of this study indicated that p38 α and EGFR might play an important role in the development and growth of lung cancer and might be a potential therapeutic target for the treatment of lung cancer.
Collapse
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Aria
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
198
|
Targeting Metabolic Bottlenecks in Lung Cancer. Trends Cancer 2019; 5:457-459. [PMID: 31421901 DOI: 10.1016/j.trecan.2019.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/14/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022]
Abstract
Lung cancer remains one of the most genetically complex, aggressive, and lethal solid malignancies. Understanding how distinct lung cancer mutations give rise to altered nutrient requirements and promote immune evasion in the context of a heterogeneous lung tumor microenvironment is vital for the development of novel personalized therapeutic strategies.
Collapse
|
199
|
FBXO22 mediates polyubiquitination and inactivation of LKB1 to promote lung cancer cell growth. Cell Death Dis 2019; 10:486. [PMID: 31217475 PMCID: PMC6584689 DOI: 10.1038/s41419-019-1732-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/19/2019] [Accepted: 05/27/2019] [Indexed: 12/19/2022]
Abstract
Liver kinase B1 (LKB1) regulates both cell growth and energy metabolism. Inactivated mutations of LKB1, observed in 20–30% of nonsmall cell lung cancers (NSCLC), contribute significantly to lung cancer malignancy progression. However, the upstream signalings regulating LKB1 activity remain incompletely understood. Here, we present evidence that FBXO22 interacts with and promotes polyubiquitination of LKB1. More intriguingly, FBXO22 mediates Lys-63-linked LKB1 polyubiquitination and inhibits kinase activity of LKB1. Furthermore, over-expression of FBXO22 promotes NSCLC cell growth through inhibiting LKB1-AMPK-mTOR signaling in vitro and in vivo. Clinically, FBXO22 is highly expressed in human lung adenocarcinoma and high FBXO22 expression predicts significant poor prognosis. Our study provides new insights into the upstream regulation of LKB1 activation and identifies FBXO22 as a potential therapeutic target for lung cancer treatment.
Collapse
|
200
|
Rabin-Court A, Rodrigues MR, Zhang XM, Perry RJ. Obesity-associated, but not obesity-independent, tumors respond to insulin by increasing mitochondrial glucose oxidation. PLoS One 2019; 14:e0218126. [PMID: 31188872 PMCID: PMC6561592 DOI: 10.1371/journal.pone.0218126] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/27/2019] [Indexed: 12/17/2022] Open
Abstract
Obesity is associated with increased incidence and worse prognosis of more than one dozen tumor types; however, the molecular mechanisms for this association remain under debate. We hypothesized that insulin, which is elevated in obesity-driven insulin resistance, would increase tumor glucose oxidation in obesity-associated tumors. To test this hypothesis, we applied and validated a stable isotope method to measure the ratio of pyruvate dehydrogenase flux to citrate synthase flux (VPDH/VCS, i.e. the percent of total mitochondrial oxidation fueled by glucose) in tumor cells. Using this method, we found that three tumor cell lines associated with obesity (colon cancer [MC38], breast cancer [4T1], and prostate cancer [TRAMP-C3] cells) increase VPDH/VCS in response to physiologic concentrations of insulin. In contrast, three tumor cell lines that are not associated with obesity (melanoma [YUMM1.7], B cell lymphoma [BCL1 clone 5B1b], and small cell lung cancer [NCI-H69] cells) exhibited no oxidative response to insulin. The observed increase in glucose oxidation in response to insulin correlated with a dose-dependent increase in cell division in obesity-associated tumor cell lines when grown in insulin, whereas no alteration in cell division was seen in tumor types not associated with obesity. These data reveal that a shift in substrate preference in the setting of physiologic insulin may comprise a metabolic signature of obesity-associated tumors that differs from that of those not associated with obesity.
Collapse
Affiliation(s)
- Aviva Rabin-Court
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Marcos R. Rodrigues
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xian-Man Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Rachel J. Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|