1
|
Hajaj E, Pozzi S, Erez A. From the Inside Out: Exposing the Roles of Urea Cycle Enzymes in Tumors and Their Micro and Macro Environments. Cold Spring Harb Perspect Med 2024; 14:a041538. [PMID: 37696657 PMCID: PMC10982720 DOI: 10.1101/cshperspect.a041538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Catabolic pathways change in anabolic diseases such as cancer to maintain metabolic homeostasis. The liver urea cycle (UC) is the main catabolic pathway for disposing excess nitrogen. Outside the liver, the UC enzymes are differentially expressed based on each tissue's needs for UC intermediates. In tumors, there are changes in the expression of UC enzymes selected for promoting tumorigenesis by increasing the availability of essential UC substrates and products. Consequently, there are compensatory changes in the expression of UC enzymes in the cells that compose the tumor microenvironment. Moreover, extrahepatic tumors induce changes in the expression of the liver UC, which contribute to the systemic manifestations of cancer, such as weight loss. Here, we review the multilayer changes in the expression of UC enzymes throughout carcinogenesis. Understanding the changes in UC expression in the tumor and its micro and macro environment can help identify biomarkers for early cancer diagnosis and vulnerabilities that can be targeted for therapy.
Collapse
Affiliation(s)
- Emma Hajaj
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sabina Pozzi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ayelet Erez
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
2
|
Gao P, Mei Z, Liu Z, Zhu D, Yuan H, Zhao R, Xu K, Zhang T, Jiang Y, Suo C, Chen X. Association between serum urea concentrations and the risk of colorectal cancer, particularly in individuals with type 2 diabetes: A cohort study. Int J Cancer 2024; 154:297-306. [PMID: 37671773 DOI: 10.1002/ijc.34719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/07/2023]
Abstract
Dysregulation of the urea cycle (UC) has been detected in colorectal cancer (CRC). However, the impact of the UC's end product, urea, on CRC development remains unclear. We investigated the association between serum urea and CRC risk based on the data of 348 872 participants cancer-free at recruitment from the UK Biobank. Multivariable Cox proportional hazards models were fitted to conduct risk estimates. Stratification analyses based on sex, diet pattern, metabolic factors (including body mass index [BMI], the estimated glomerular filtration rate [eGFR] and type 2 diabetes [T2D]) and genetic profiles (the polygenic risk score [PRS] of CRC) were conducted to find potential modifiers. During an average of 9.0 years of follow-up, we identified 3408 (1.0%) CRC incident cases. Serum urea showed a nonlinear relationship with CRC risk (P-nonlinear: .035). Lower serum urea levels were associated with a higher CRC risk, with a fully-adjusted hazard ratio (HR) of 1.26 (95% confidence interval [CI]: 1.13-1.41) in the first quartile (Q1) of urea, compared to the Q4. This association was largely consistent across subgroups of sex, protein diet, BMI, eGFR and CRC-PRSs (P-interaction >.05); however, it was stronger in the T2D, with an interaction between urea and T2D on both additive (synergy index: 3.32, [95% CI: 1.24-8.88]) and multiplicative scales (P-interaction: .019). Lower serum urea concentrations were associated with an increased risk of CRC, with a more pronounced effect observed in individuals with T2D. Maintaining stable levels of serum urea has important implications for CRC prevention, particularly in individuals with T2D.
Collapse
Affiliation(s)
- Peipei Gao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Zhendong Mei
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Dongliang Zhu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Renjia Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Kelin Xu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China
| |
Collapse
|
3
|
Gupta S, Westacott MJ, Ayers DG, Weiss SJ, Whitley P, Mueller C, Weaver DC, Schneider DJ, Karimpour-Fard A, Hunter LE, Drolet DW, Janjic N. Plasma proteome of growing tumors. Sci Rep 2023; 13:12195. [PMID: 37500700 PMCID: PMC10374562 DOI: 10.1038/s41598-023-38079-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/03/2023] [Indexed: 07/29/2023] Open
Abstract
Early detection of cancer is vital for the best chance of successful treatment, but half of all cancers are diagnosed at an advanced stage. A simple and reliable blood screening test applied routinely would therefore address a major unmet medical need. To gain insight into the value of protein biomarkers in early detection and stratification of cancer we determined the time course of changes in the plasma proteome of mice carrying transplanted human lung, breast, colon, or ovarian tumors. For protein measurements we used an aptamer-based assay which simultaneously measures ~ 5000 proteins. Along with tumor lineage-specific biomarkers, we also found 15 markers shared among all cancer types that included the energy metabolism enzymes glyceraldehyde-3-phosphate dehydrogenase, glucose-6-phophate isomerase and dihydrolipoyl dehydrogenase as well as several important biomarkers for maintaining protein, lipid, nucleotide, or carbohydrate balance such as tryptophanyl t-RNA synthetase and nucleoside diphosphate kinase. Using significantly altered proteins in the tumor bearing mice, we developed models to stratify tumor types and to estimate the minimum detectable tumor volume. Finally, we identified significantly enriched common and unique biological pathways among the eight tumor cell lines tested.
Collapse
Affiliation(s)
- Shashi Gupta
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO, 80301, USA
| | | | - Deborah G Ayers
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO, 80301, USA
| | - Sophie J Weiss
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO, 80301, USA
| | - Penn Whitley
- Boulder BioConsulting, Inc., 325 S 68th St., Boulder, CO, 80303, USA
| | | | - Daniel C Weaver
- Boulder BioConsulting, Inc., 325 S 68th St., Boulder, CO, 80303, USA
| | | | - Anis Karimpour-Fard
- University of Colorado School of Medicine, Mailstop 8303, Aurora, CO, 80045, USA
| | - Lawrence E Hunter
- University of Colorado School of Medicine, Mailstop 8303, Aurora, CO, 80045, USA
| | - Daniel W Drolet
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO, 80301, USA
| | - Nebojsa Janjic
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO, 80301, USA.
| |
Collapse
|
4
|
Zhao Y, Chen Y, Wei L, Ran J, Wang K, Zhu S, Liu Q. p53 inhibits the Urea cycle and represses polyamine biosynthesis in glioma cell lines. Metab Brain Dis 2023; 38:1143-1153. [PMID: 36745250 DOI: 10.1007/s11011-023-01173-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/18/2023] [Indexed: 02/07/2023]
Abstract
Glioma is the most common malignant tumor of the central nervous system. The urea cycle (UC) is an essential pathway to convert excess nitrogen and ammonia into the less toxic urea in humans. However, less is known about the functional significance of the urea cycle in glioma. p53 functions as a tumor suppressor and modulates several cellular functions and disease processes. In the present study, we aimed to explore whether p53 influences glioma progression by regulating the urea cycle. Here, we demonstrated the inhibitory impact of p53 on the expression of urea cycle enzymes and urea genesis in glioma cells. The level of polyamine, a urea cycle metabolite, was also regulated by p53 in glioma cells. Carbamoyl phosphate synthetase-1 (CPS1) is the first key enzyme involved in the urea cycle. Functionally, we demonstrated that CPS1 knockdown suppressed glioma cell proliferation, migration and invasion. Mechanistically, we demonstrated that the expression of ornithine decarboxylase (ODC), which determines the generation of polyamine, was regulated by CPS1. In addition, the impacts of p53 knockdown on ODC expression, glioma cell growth and aggressive phenotypes were significantly reversed by CPS1 inhibition. In conclusion, these results demonstrated that p53 inhibits polyamine metabolism by suppressing the urea cycle, which inhibits glioma progression.
Collapse
Affiliation(s)
- Yuhong Zhao
- Institute of Neuroscience, Chongqing Medical University Basic Medical College, Chongqing, 400016, China
| | - Yingxi Chen
- Department of basic Medicine, Chongqing College of traditional Chinese Medicine, Chongqing, 402760, PR China
| | - Ling Wei
- Institute of Neuroscience, Chongqing Medical University Basic Medical College, Chongqing, 400016, China
| | - Jianhua Ran
- Institute of Neuroscience, Chongqing Medical University Basic Medical College, Chongqing, 400016, China
| | - Kejian Wang
- Institute of Neuroscience, Chongqing Medical University Basic Medical College, Chongqing, 400016, China
| | - Shujuan Zhu
- Institute of Neuroscience, Chongqing Medical University Basic Medical College, Chongqing, 400016, China
| | - Qian Liu
- Institute of Neuroscience, Chongqing Medical University Basic Medical College, Chongqing, 400016, China.
- Department of basic Medicine, Chongqing College of traditional Chinese Medicine, Chongqing, 402760, PR China.
| |
Collapse
|
5
|
Khare S, Kim LC, Lobel G, Doulias PT, Ischiropoulos H, Nissim I, Keith B, Simon MC. ASS1 and ASL suppress growth in clear cell renal cell carcinoma via altered nitrogen metabolism. Cancer Metab 2021; 9:40. [PMID: 34861885 PMCID: PMC8642968 DOI: 10.1186/s40170-021-00271-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/08/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Kidney cancer is a common adult malignancy in the USA. Clear cell renal cell carcinoma (ccRCC), the predominant subtype of kidney cancer, is characterized by widespread metabolic changes. Urea metabolism is one such altered pathway in ccRCC. The aim of this study was to elucidate the contributions of urea cycle enzymes, argininosuccinate synthase 1 (ASS1), and argininosuccinate lyase (ASL) towards ccRCC progression. METHODS We employed a combination of computational, genetic, and metabolomic tools along with in vivo animal models to establish a tumor-suppressive role for ASS1 and ASL in ccRCC. RESULTS We show that the mRNA and protein expression of urea cycle enzymes ASS1 and ASL are reduced in ccRCC tumors when compared to the normal kidney. Furthermore, the loss of ASL in HK-2 cells (immortalized renal epithelial cells) promotes growth in 2D and 3D growth assays, while combined re-expression of ASS1 and ASL in ccRCC cell lines suppresses growth in 2D, 3D, and in vivo xenograft models. We establish that this suppression is dependent on their enzymatic activity. Finally, we demonstrate that conservation of cellular aspartate, regulation of nitric oxide synthesis, and pyrimidine production play pivotal roles in ASS1+ASL-mediated growth suppression in ccRCC. CONCLUSIONS ccRCC tumors downregulate the components of the urea cycle including the enzymes argininosuccinate synthase 1 (ASS1) and argininosuccinate lyase (ASL). These cytosolic enzymes lie at a critical metabolic hub in the cell and are involved in aspartate catabolism and arginine and nitric oxide biosynthesis. Loss of ASS1 and ASL helps cells redirect aspartate towards pyrimidine synthesis and support enhanced proliferation. Additionally, reduced levels of ASS1 and ASL might help regulate nitric oxide (NO) generation and mitigate its cytotoxic effects. Overall, our work adds to the understanding of urea cycle enzymes in a context-independent of ureagenesis, their role in ccRCC progression, and uncovers novel potential metabolic vulnerabilities in ccRCC.
Collapse
Affiliation(s)
- Sanika Khare
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Laura C Kim
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Graham Lobel
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paschalis-Thomas Doulias
- Children's Hospital of Philadelphia Research Institute and Departments of Pediatrics and Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute and Departments of Pediatrics and Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Itzhak Nissim
- Division of Genetics and Metabolism, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Biochemistry, and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Brian Keith
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Hajaj E, Sciacovelli M, Frezza C, Erez A. The context-specific roles of urea cycle enzymes in tumorigenesis. Mol Cell 2021; 81:3749-3759. [PMID: 34469752 DOI: 10.1016/j.molcel.2021.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022]
Abstract
The expression of the urea cycle (UC) proteins is dysregulated in multiple cancers, providing metabolic benefits to tumor survival, proliferation, and growth. Here, we review the main changes described in the expression of UC enzymes and metabolites in different cancers at various stages and suggest that these changes are dynamic and should hence be viewed in a context-specific manner. Understanding the evolvability in the activity of the UC pathway in cancer has implications for cancer-immune cell interactions and for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Emma Hajaj
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Marco Sciacovelli
- Medical Research Council Cancer Unit, University of Cambridge, Box 197, Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Box 197, Biomedical Campus, Cambridge CB2 0XZ, UK.
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
7
|
Cai Y, Chow JPH, Leung YO, Lu X, Yuen CH, Lee WL, Chau KC, Yang LL, Wong RMH, Lam JYT, Chow DTL, Chung SHK, Kwok SY, Leung YC. NEI-01-Induced Arginine Deprivation Has Potent Activity Against Acute Myeloid Leukemia Cells Both In Vitro and In Vivo. Mol Cancer Ther 2021; 20:2218-2227. [PMID: 34433661 DOI: 10.1158/1535-7163.mct-21-0120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/24/2021] [Accepted: 08/18/2021] [Indexed: 11/16/2022]
Abstract
Recent studies have revealed that targeting amino acid metabolic enzymes is a promising strategy in cancer therapy. Acute myeloid leukemia (AML) downregulates the expression of argininosuccinate synthase (ASS1), a recognized rate-limiting enzyme for arginine synthesis, and yet displays a critical dependence on extracellular arginine for survival and proliferation. This dependence on extracellular arginine, also known as arginine auxotrophy, suggests that arginine deprivation would be a treatment strategy for AML. NEI-01, a novel arginine-depleting enzyme, is capable of binding to serum albumin to extend its circulating half-life, leading to a potent anticancer activity. Here we reported the preclinical activity of NEI-01 in arginine auxotrophic AMLs. NEI-01 efficiently depleted arginine both in vitro and in vivo NEI-01-induced arginine deprivation was cytotoxic to arginine auxotrophic AML cells through induction of cell-cycle arrest and apoptosis. Furthermore, the potent anti-leukemia activities of NEI-01 were observed in three different types of mouse models including human cell line-derived xenograft, mouse cell line-derived homografts in syngeneic mice and patient-derived xenograft. This preclinical data provide strong evidence to support the potential use of NEI-01 as a therapeutic approach in AML treatment.
Collapse
Affiliation(s)
- Yijun Cai
- New Epsilon Innovation Limited, Hong Kong, China.
| | | | - Yu-On Leung
- New Epsilon Innovation Limited, Hong Kong, China
| | - Xiaoxu Lu
- New Epsilon Innovation Limited, Hong Kong, China
| | - Chak-Ho Yuen
- New Epsilon Innovation Limited, Hong Kong, China
| | - Wing Lun Lee
- New Epsilon Innovation Limited, Hong Kong, China
| | - Ka-Chun Chau
- New Epsilon Innovation Limited, Hong Kong, China
| | - Liz L Yang
- New Epsilon Innovation Limited, Hong Kong, China
| | | | | | | | | | - Sui-Yi Kwok
- New Epsilon Innovation Limited, Hong Kong, China.
| | - Yun-Chung Leung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology and Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
8
|
Zhang Y, Chung SF, Tam SY, Leung YC, Guan X. Arginine deprivation as a strategy for cancer therapy: An insight into drug design and drug combination. Cancer Lett 2021; 502:58-70. [PMID: 33429005 DOI: 10.1016/j.canlet.2020.12.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022]
Abstract
Extensive studies have shown that cancer cells have specific nutrient auxotrophy and thus have much a higher demand for certain nutrients than normal cells. Amino acid deprivation has attracted much attention in cancer therapy with positive outcomes from clinical trials. Arginine, as one of the conditionally essential amino acids, plays a pivotal role in cellular division and metabolism. Since many types of cancer cells exhibit decreased expression of argininosuccinate synthetase and/or ornithine transcarbamylase, they are auxotrophic for arginine, which makes arginine deprivation an accessible choice for cancer treatment. Arginine deiminase (ADI) and human arginase (hArg) are the two major protein drugs used for arginine deprivation and are undergoing many clinical trials. However, the clinical application of ADI and hArg is facing some common problems, including their short half-lives, immunogenicity and inconsistent production, which underlines the importance of improving these drugs using protein engineering techniques. Thus, we systematically review the latest studies of protein engineering and anti-cancer studies based on in vitro, in vivo and clinical models of ADI and hArg, and we include the latest studies on drug combinations consisting of ADI/hArg with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Yu Zhang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China; Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Center for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China; Shanghai Engineering Research Center for Food Rapid Detection, Shanghai, China
| | - Sai-Fung Chung
- Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Center for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Suet-Ying Tam
- Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Center for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Yun-Chung Leung
- Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Center for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
| | - Xiao Guan
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China.
| |
Collapse
|
9
|
Guan F, Kang Z, Zhang JT, Xue NN, Yin H, Wang L, Mao BB, Peng WC, Zhang BL, Liang X, Hu ZQ. KLF7 promotes polyamine biosynthesis and glioma development through transcriptionally activating ASL. Biochem Biophys Res Commun 2019; 514:51-57. [DOI: 10.1016/j.bbrc.2019.04.120] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 04/13/2019] [Accepted: 04/16/2019] [Indexed: 11/26/2022]
|
10
|
Gong R, He L, Zhou H, Cheng S, Ren F, Chen J, Ren J. Down-regulation of argininosuccinate lyase induces hepatoma cell apoptosis through activating Bax signaling pathway. Genes Dis 2018; 6:296-303. [PMID: 32042869 PMCID: PMC6997574 DOI: 10.1016/j.gendis.2018.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/12/2018] [Indexed: 01/11/2023] Open
Abstract
Argininosuccinate lyase (ASL) plays an important role in the hepatic urea cycle, and can catalyze the reversible reaction of argininosuccinate to arginine and fumarate. However, the function of ASL in hepatocellular carcinoma (HCC) is not fully understood. In this study, we found that ASL expression was frequently upregulated in HCC tissues and HCC cell lines. Knock down of ASL inhibited cell proliferation and induced apoptosis in HCC cells. Mechanistic studies revealed the BCL2-associated X protein (Bax) signaling pathway which determines cancer cell apoptosis was regulated by ASL. Moreover, the depletion of Bax restored the inhibition of cell growth and reduced apoptosis initiated by ASL silencing. Together, the study demonstrated that ASL regulated HCC cell growth and apoptosis by modulating Bax signaling. Thus, the therapeutic targeting of ASL may offer options for HCC treatment.
Collapse
Affiliation(s)
- Rui Gong
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Lin He
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - HongZhong Zhou
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - ShengTao Cheng
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Fang Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Juan Chen
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - JiHua Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
11
|
Abstract
Cancer cells reprogramme metabolism to maximize the use of nitrogen and carbon for the anabolic synthesis of macromolecules that are required during tumour proliferation and growth. To achieve this aim, one strategy is to reduce catabolism and nitrogen disposal. The urea cycle (UC) in the liver is the main metabolic pathway to convert excess nitrogen into disposable urea. Outside the liver, UC enzymes are differentially expressed, enabling the use of nitrogen for the synthesis of UC intermediates that are required to accommodate cellular needs. Interestingly, the expression of UC enzymes is altered in cancer, revealing a revolutionary mechanism to maximize nitrogen incorporation into biomass. In this Review, we discuss the metabolic benefits underlying UC deregulation in cancer and the relevance of these alterations for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Peter Szlosarek
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK
- Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Arkaitz Carracedo
- CIC bioGUNE, Bizkaia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country, Bilbao, Spain
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
12
|
Keshet R, Erez A. Arginine and the metabolic regulation of nitric oxide synthesis in cancer. Dis Model Mech 2018; 11:11/8/dmm033332. [PMID: 30082427 PMCID: PMC6124554 DOI: 10.1242/dmm.033332] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nitric oxide (NO) is a signaling molecule that plays important roles in diverse biological processes and thus its dysregulation is involved in the pathogenesis of various disorders. In cancer, NO has broad and sometimes dichotomous roles; it is involved in cancer initiation and progression, but also restricts cancer proliferation and invasion, and contributes to the anti-tumor immune response. The importance of NO in a range of cellular processes is exemplified by its tight spatial and dosage control at multiple levels, including via its transcriptional, post-translational and metabolic regulation. In this Review, we focus on the regulation of NO via the synthesis and availability of its precursor, arginine, and discuss the implications of this metabolic regulation for cancer biology and therapy. Despite the established contribution of NO to cancer pathogenesis, the implementation of NO-related cancer therapeutics remains limited, likely due to the challenge of targeting and inducing its protective functions in a cell- and dosage-specific manner. A better understanding of how arginine regulates the production of NO in cancer might thus support the development of anti-cancer drugs that target this key metabolic pathway, and other metabolic pathways involved in NO production.
Collapse
Affiliation(s)
- Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
13
|
Affiliation(s)
- Wissam Zam
- Al-Andalus University for Medical Sciences, Syrian Arab Republic
| |
Collapse
|
14
|
Hung YH, Huang HL, Chen WC, Yen MC, Cho CY, Weng TY, Wang CY, Chen YL, Chen LT, Lai MD. Argininosuccinate lyase interacts with cyclin A2 in cytoplasm and modulates growth of liver tumor cells. Oncol Rep 2016; 37:969-978. [PMID: 28035420 PMCID: PMC5355748 DOI: 10.3892/or.2016.5334] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/13/2016] [Indexed: 12/11/2022] Open
Abstract
Arginine is a critical amino acid in specific cancer types including hepatocellular carcinoma (HCC) and melanoma. Novel molecular mechanisms and therapeutic targets in arginine metabolism-mediated cancer formation await further identification. Our laboratory has previously demonstrated that arginine metabolic enzyme argininosuccinate lyase (ASL) promoted HCC formation in part via maintenance of cyclin A2 protein expression and arginine production for channeling to nitric oxide synthase. In this study, we investigated the mechanism by which ASL regulates cyclin A2 expression. We found that ASL interacted with cyclin A2 in HCC cells and the localization of their interaction was in the cytoplasm. Mutation of essential residues for enzymatic activity of ASL did not affect the binding of ASL to cyclin A2. Moreover, the mutant ASL retained the ability to restore the decreased tumorigenicity caused by ASL shRNA. Furthermore, overexpression of ASL conferred resistance to arginine deprivation therapy. Finally, the important pathways and potential therapeutic targets in ASL-regulated HCC were identified by bioinformatics analyses with Metacore database and Connectivity Map database. Our analyses suggested that bisoprolol, celecoxib, and ipratropium bromide, are potential therapeutics for ASL-regulated HCC formation. Thus, ASL interacts with cyclin A2 in cytoplasm, and may promote HCC formation through this non-enzymatic function. Overexpression of ASL may be a contributing factor in drug resistance for arginine deprivation therapy.
Collapse
Affiliation(s)
- Yu-Hsuan Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Hau-Lun Huang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| | - Wei-Ching Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Chien-Yu Cho
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Tzu-Yang Weng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Chih-Yang Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Yi-Ling Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan, R.O.C
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 701, Taiwan, R.O.C
| | - Ming-Derg Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| |
Collapse
|
15
|
Albaugh VL, Pinzon-Guzman C, Barbul A. Arginine-Dual roles as an onconutrient and immunonutrient. J Surg Oncol 2016; 115:273-280. [PMID: 27861915 DOI: 10.1002/jso.24490] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/22/2016] [Indexed: 12/12/2022]
Abstract
Arginine is an important player in numerous biologic processes and studies have demonstrated its importance for cellular growth that becomes limiting in states of rapid turnover (e.g., malignancy). Thus, arginine deprivation therapy is being examined as an adjuvant cancer therapy, however, arginine is also necessary for immune destruction of malignant cells. Herein we review the data supporting arginine deprivation or supplementation in cancer treatment and the currently registered trials aimed at understanding these divergent strategies. J. Surg. Oncol. 2017;115:273-280. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vance L Albaugh
- Division of General Surgery, Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Carolina Pinzon-Guzman
- Division of General Surgery, Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Adrian Barbul
- Division of General Surgery, Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
16
|
Huang HL, Chen WC, Hsu HP, Cho CY, Hung YH, Wang CY, Lai MD. Silencing of argininosuccinate lyase inhibits colorectal cancer formation. Oncol Rep 2016; 37:163-170. [DOI: 10.3892/or.2016.5221] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/14/2016] [Indexed: 11/05/2022] Open
|
17
|
Chen WC, Chang YS, Hsu HP, Yen MC, Huang HL, Cho CY, Wang CY, Weng TY, Lai PT, Chen CS, Lin YJ, Lai MD. Therapeutics targeting CD90-integrin-AMPK-CD133 signal axis in liver cancer. Oncotarget 2016; 6:42923-37. [PMID: 26556861 PMCID: PMC4767481 DOI: 10.18632/oncotarget.5976] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/12/2015] [Indexed: 12/28/2022] Open
Abstract
CD90 is used as a marker for cancer stem cell in liver cancer. We aimed to study the mechanism by which CD90 promoted liver cancer progression and identify the new therapeutic targets on CD90 signal pathway. Ectopic expression of CD90 in liver cancer cell lines enhanced anchorage-independent growth and tumor progression. Furthermore, CD90 promoted sphere formation in vitro and upregulated the expression of the cancer stem cell marker CD133. The CD133 expression was higher in CD45-CD90+ cells in liver cancer specimen. The natural carcinogenic molecules TGF-β-1, HGF, and hepatitis B surface antigen increased the expression of CD90 and CD133. Inhibition of CD90 by either shRNA or antibody attenuated the induction of CD133 and anchorage-independent growth. Lentiviral delivery of CD133 shRNA abolished the tumorigenicity induced by CD90. Ectopic expression of CD90 induced mTOR phosphorylation and AMPK dephosphorylation. Mutation of integrin binding-RLD domain in CD90 attenuated the induction of CD133 and anchorage-independent growth. Similar results were observed after silencing β3 integrin. Signaling analyses revealed that AMPK/mTOR and β3 integrin were required for the induction of CD133 and tumor formation by CD90. Importantly, the energy restriction mimetic agent OSU-CG5 reduced the CD90 population in fresh liver tumor sample and repressed the tumor growth. In contrast, sorafenib did not decrease the CD90+ population. In conclusion, the signal axis of CD90-integrin-mTOR/AMPK-CD133 is critical for promoting liver carcinogenesis. Molecules inhibiting the signal axis, including OSU-CG5 and other inhibitors, may serve as potential novel cancer therapeutic targets in liver cancer.
Collapse
Affiliation(s)
- Wei-Ching Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yung-Sheng Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Chi Yen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hau-Lun Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Yu Cho
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Yang Weng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Ting Lai
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Ching-Shih Chen
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yih-Jyh Lin
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center for Infectious Diseases and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
18
|
Role of the anti-glioma drug AT13148 in the inhibition of Notch signaling pathway. Gene 2015; 573:153-9. [DOI: 10.1016/j.gene.2015.07.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 02/03/2015] [Accepted: 07/11/2015] [Indexed: 11/18/2022]
|
19
|
Huang HL, Chen WC, Hsu HP, Cho CY, Hung YH, Wang CY, Lai MD. Argininosuccinate lyase is a potential therapeutic target in breast cancer. Oncol Rep 2015; 34:3131-9. [PMID: 26397737 DOI: 10.3892/or.2015.4280] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/23/2015] [Indexed: 11/06/2022] Open
Abstract
Arginine is a non-essential amino acid that modulates nitric oxide production and cancer homeostasis. In our previous study, we observed that blocking argininosuccinate lyase (ASL) attenuates tumor progression in liver cancer. However, the role of ASL in human breast cancer has been studied to a lesser degree. In the present study, we investigated the effect of targeting ASL in breast cancer. We found that ASL was induced by ER stress and was significantly upregulated in breast cancer tissues compared to that in the corresponding normal tissues. Downregulation of ASL inhibited the growth of breast cancer in vitro and in vivo. The level of cell cycle-related gene, cyclin A2, was reduced and was accompanied by a delay in G2/M transition. ASL shRNA-induced cell inhibition was rescued by exogenous cyclin A2. Furthermore, autophagy was observed in the cells expressing ASL shRNA, and inhibition of autophagy reduced cell growth, indicating that autophagy played a cell survival role in the ASL knockdown cells. Moreover, inhibition of ASL reduced NO content. Introduction of the NO donor partially restored the growth inhibition by ASL shRNA. Thus, the mechanism induced by ASL shRNA which occurred in human breast cancer may be attributed to a decrease in cyclin A2 and NO.
Collapse
Affiliation(s)
- Hau-Lun Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Wei-Ching Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Hui-Ping Hsu
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Chien-Yu Cho
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Yu-Hsuan Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Chih-Yang Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| | - Ming-Derg Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan, R.O.C
| |
Collapse
|
20
|
Bogdan C. Nitric oxide synthase in innate and adaptive immunity: an update. Trends Immunol 2015; 36:161-78. [PMID: 25687683 DOI: 10.1016/j.it.2015.01.003] [Citation(s) in RCA: 586] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/14/2015] [Accepted: 01/14/2015] [Indexed: 12/22/2022]
Abstract
Thirty years after the discovery of its production by activated macrophages, our appreciation of the diverse roles of nitric oxide (NO) continues to grow. Recent findings have not only expanded our understanding of the mechanisms controlling the expression of NO synthases (NOS) in innate and adaptive immune cells, but have also revealed new functions and modes of action of NO in the control and escape of infectious pathogens, in T and B cell differentiation, and in tumor defense. I discuss these findings, in the context of a comprehensive overview of the various sources and multiple reaction partners of NO, and of the regulation of NOS2 by micromilieu factors, antisense RNAs, and 'unexpected' cytokines.
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie, und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Wasserturmstraße 3/5, 91054 Erlangen, Germany.
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW There has been an increased and renewed interest in metabolic therapy for cancer, particularly Arg deprivation. The purpose of this review is to highlight recent studies that focus on Arg-dependent malignancies with Arginine (Arg)-degrading enzymes, including arginase and Arg deiminase. RECENT FINDINGS New developments in this area include understanding of the role of most significantly downregulated gene regulating amino acid metabolism, argininosuccinate synthetase and its expression and therapeutic relevance in different tumors. Recent studies have also shed light on the mechanism of tumor cell death with Arg deprivation, with arginase and pegylated Arg deiminase. Particularly important is understanding the mechanism of resistance that cancers develop after such drug exposure. Finally, recent clinical trials have been performed or are ongoing to use Arg deprivation as treatment for advanced malignancies. SUMMARY Arg deprivation is a promising approach for the treatment of various malignancies.
Collapse
Affiliation(s)
- Lynn G Feun
- aSylvester Comprehensive Cancer Center, University of Miami, Miami, Florida bDepartment of Translational Molecular Pathology, The University of Texas MD, Anderson Cancer Center, Houston, Texas cSylvester Comprehensive Cancer Center, University of Miami, VA Medical Center, Miami, Florida, USA
| | | | | |
Collapse
|
22
|
HUNG YUHSUAN, CHAN YISHIN, CHANG YUNGSHENG, LEE KUOTING, HSU HUIPING, YEN MENGCHI, CHEN WEICHING, WANG CHIHYANG, LAI MINGDERG. Fatty acid metabolic enzyme acyl-CoA thioesterase 8 promotes the development of hepatocellular carcinoma. Oncol Rep 2014; 31:2797-803. [DOI: 10.3892/or.2014.3155] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 03/20/2014] [Indexed: 11/06/2022] Open
|
23
|
Phillips MM, Sheaff MT, Szlosarek PW. Targeting arginine-dependent cancers with arginine-degrading enzymes: opportunities and challenges. Cancer Res Treat 2013; 45:251-62. [PMID: 24453997 PMCID: PMC3893322 DOI: 10.4143/crt.2013.45.4.251] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/13/2013] [Indexed: 12/13/2022] Open
Abstract
Arginine deprivation is a novel antimetabolite strategy for the treatment of arginine-dependent cancers that exploits differential expression and regulation of key urea cycle enzymes. Several studies have focused on inactivation of argininosuccinate synthetase 1 (ASS1) in a range of malignancies, including melanoma, hepatocellular carcinoma (HCC), mesothelial and urological cancers, sarcomas, and lymphomas. Epigenetic silencing has been identified as a key mechanism for loss of the tumor suppressor role of ASS1 leading to tumoral dependence on exogenous arginine. More recently, dysregulation of argininosuccinate lyase has been documented in a subset of arginine auxotrophic glioblastoma multiforme, HCC and in fumarate hydratase-mutant renal cancers. Clinical trials of several arginine depletors are ongoing, including pegylated arginine deiminase (ADI-PEG20, Polaris Group) and bioengineered forms of human arginase. ADI-PEG20 is furthest along the path of clinical development from combinatorial phase 1 to phase 3 trials and is described in more detail. The challenge will be to identify tumors sensitive to drugs such as ADI-PEG20 and integrate these agents into multimodality drug regimens using imaging and tissue/fluid-based biomarkers as predictors of response. Lastly, resistance pathways to arginine deprivation require further study to optimize arginine-targeted therapies in the oncology clinic.
Collapse
Affiliation(s)
- Melissa M. Phillips
- Center for Molecular Oncology, Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
- St Bartholomew's Hospital, London, UK
| | - Michael T. Sheaff
- Pathology Group, Institute of Cell and Molecular Sciences, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Peter W. Szlosarek
- Center for Molecular Oncology, Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
- St Bartholomew's Hospital, London, UK
| |
Collapse
|