1
|
Abo Qoura L, Balakin KV, Hoffman RM, Pokrovsky VS. The potential of methioninase for cancer treatment. Biochim Biophys Acta Rev Cancer 2024; 1879:189122. [PMID: 38796027 DOI: 10.1016/j.bbcan.2024.189122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
Cancer cells are addicted to L-methionine (L-Met) and have a much greater requirement for L-Met than normal cells due to excess transmethylation, termed the Hoffman effect. By targeting this vulnerability through dietary restriction of L-Met, researchers have been able to achieve promising results in inhibiting tumor growth and eradicating cancer cells. Methioninase (EC 4.4.1.11; METase) catalyzes the transformation of L-Met into α-ketobutyrate, ammonia, and methanethiol. The use of METase was initially limited due to its poor stability in vivo, high immunogenicity, and enzyme-induced inactivating antibodies. These issues could be partially resolved by PEGylation, encapsulation in erythrocytes, and various site-directed mutagenesis. The big breakthrough came when it was discovered that METase is effectively administered orally. The enzyme L-asparaginase is approved by the FDA for treatment of acute lymphoblastic leukemia. METase has more potential as a therapeutic since addiction to L-Met is a general and fundamental hallmark of cancer.
Collapse
Affiliation(s)
- Louay Abo Qoura
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), 117198 Moscow, Russia; N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, 115478 Moscow, Russia
| | | | - Robert M Hoffman
- AntiCancer Inc., San Diego, CA 92111, USA; Department of Surgery, University of California, San Diego, La Jolla, CA 92037-7400, USA
| | - Vadim S Pokrovsky
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), 117198 Moscow, Russia; N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, 115478 Moscow, Russia.
| |
Collapse
|
2
|
Wei X, Chow HY, Chong HC, Leung SL, Ho MK, Lee MY, Leung YC. Arginine Is a Novel Drug Target for Arginine Decarboxylase in Human Colorectal Cancer Cells. Int J Mol Sci 2023; 24:13741. [PMID: 37762044 PMCID: PMC10531272 DOI: 10.3390/ijms241813741] [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: 07/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) has been proven to be highly reliant on arginine availability. Limiting arginine-rich foods or treating patients with arginine-depleting enzymes arginine deiminase (ADI) or arginase can suppress colon cancer. However, arginase and ADI are not the best drug candidates for CRC. Ornithine, the product of arginase, can enhance the supply of polyamine, which favors CRC cell growth, while citrulline, the product of ADI, faces the problem of arginine recycling due to the overexpression of argininosuccinate synthetase (ASS). Biosynthetic arginine decarboxylase (ADC), an enzyme that catalyzes the conversion of arginine to agmatine and carbon dioxide, may be a better choice as it combines both arginine depletion and suppression of intracellular polyamine synthesis via its product agmatine. ADC has anti-tumor potential yet has received much less attention than the other two arginine-depleting enzymes. In order to gain a better understanding of ADC, the preparation and the anti-cancer properties of this enzyme were explored in this study. When tested in vitro, ADC inhibited the proliferation of three colorectal cancer cell lines regardless of their ASS cellular expression. In contrast, ADC had a lesser cytotoxic effect on the human foreskin fibroblasts and rat primary hepatocytes. Further in vitro studies revealed that ADC induced S and G2/M phase cell-cycle arrest and apoptosis in HCT116 and LoVo cells. ADC-induced apoptosis in HCT116 cells followed the mitochondrial apoptotic pathway and was caspase-3-dependent. With all results obtained, we suggest that arginine is a potential target for treating colorectal cancer with ADC, and the anti-cancer properties of ADC should be more deeply investigated in the future.
Collapse
Affiliation(s)
- Xinlei Wei
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Ho-Yin Chow
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Hiu-Chi Chong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Siu-Lun Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Mei-Ki Ho
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Man-Yuen Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- 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, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
3
|
Hussain A, Xie L, Deng G, Kang X. Common alterations in plasma free amino acid profiles and gut microbiota-derived tryptophan metabolites of five types of cancer patients. Amino Acids 2023; 55:1189-1200. [PMID: 37490156 DOI: 10.1007/s00726-023-03308-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
Amino acids not only play a vital role in the synthesis of biological molecules such as proteins in cancer malignant cells, they are also essential metabolites for immune cell activation and antitumor effects in the tumor microenvironment. The abnormal changes in amino acid metabolism are closely related to the occurrence and development of tumors and immunity. Intestinal microorganisms play an essential role in amino acid metabolism, and tryptophan and its intestinal microbial metabolites are typical representatives. However, it is known that the cyclic amino acid profile is affected by specific cancer types, so relevant studies mainly focus on one type of cancer and rarely study different cancer forms at the same time. The objective of this study was to examine the PFAA profile of five cancer patients and the characteristics of tryptophan intestinal microbial metabolites to determine whether there are general amino acid changes across tumors. Plasma samples were collected from esophageal (n = 53), lung (n = 73), colorectal (n = 94), gastric (n = 55), breast cancer (n = 25), and healthy control (HC) (n = 139) subjects. PFAA profile and tryptophan metabolites were measured, and their perioperative changes were examined using high-performance liquid chromatography. Univariate analysis revealed significant differences between cancer patients and HC. Furthermore, multivariate analysis discriminated cancer patients from HC. Regression diagnosis models were established for each cancer group using differential amino acids from univariate analysis. Receiver-operating characteristic analysis was applied to evaluate these diagnosis models. Finally, GABA, arginine, tryptophan, taurine, glutamic acid, and melatonin showed common alterations across all types of cancer patients. Metabolic pathway analysis shows that the most significant enrichment pathways were tryptophan, arginine, and proline metabolism. This study provides evidence that common alterations of the metabolites mentioned above suggest their role in the pathogenesis of each cancer patient. It was suggested that multivariate models based on PFAA profiles and tryptophan metabolites might be applicable in the screening of cancer patients.
Collapse
Affiliation(s)
- Ahad Hussain
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Child Development and Learning Science of Ministry of Education of China, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Li Xie
- College of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, 210038, Jiangsu, China
| | - Guozhe Deng
- Key Laboratory of Child Development and Learning Science of Ministry of Education of China, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xuejun Kang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
- Key Laboratory of Child Development and Learning Science of Ministry of Education of China, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
4
|
Pokrovsky VS, Abo Qoura L, Morozova E, Bunik VI. Predictive markers for efficiency of the amino-acid deprivation therapies in cancer. Front Med (Lausanne) 2022; 9:1035356. [PMID: 36405587 PMCID: PMC9669297 DOI: 10.3389/fmed.2022.1035356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Amino acid deprivation therapy (AADT) is a promising strategy for developing novel anticancer treatments, based on variations in metabolism of healthy and malignant cells. L-asparaginase was the first amino acid-degrading enzyme that received FDA approval for the treatment of acute lymphoblastic leukemia (ALL). Arginase and arginine deiminase were effective in clinical trials for the treatment of metastatic melanomas and hepatocellular carcinomas. Essential dependence of certain cancer cells on methionine explains the anticancer efficacy of methionine-g-lyase. Along with significant progress in identification of metabolic vulnerabilities of cancer cells, new amino acid-cleaving enzymes appear as promising agents for cancer treatment: lysine oxidase, tyrosine phenol-lyase, cysteinase, and phenylalanine ammonia-lyase. However, sensitivity of specific cancer cell types to these enzymes differs. Hence, search for prognostic and predictive markers for AADT and introduction of the markers into clinical practice are of great importance for translational medicine. As specific metabolic pathways in cancer cells are determined by the enzyme expression, some of these enzymes may define the sensitivity to AADT. This review considers the known predictors for efficiency of AADT, emphasizing the importance of knowledge on cancer-specific amino acid significance for such predictions.
Collapse
Affiliation(s)
- Vadim S. Pokrovsky
- Laboratory of Experimental Oncology, Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), Moscow, Russia
- Laboratory of Combined Treatment, N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia
- Department of Biotechnology, Sirius University of Science and Technology, Sochi, Russia
- *Correspondence: Vadim S. Pokrovsky,
| | - Louay Abo Qoura
- Laboratory of Experimental Oncology, Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), Moscow, Russia
| | - Elena Morozova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Victoria I. Bunik
- A.N. Belozersky Institute of Physicochemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
5
|
Carpentier J, Pavlyk I, Mukherjee U, Hall PE, Szlosarek PW. Arginine Deprivation in SCLC: Mechanisms and Perspectives for Therapy. LUNG CANCER (AUCKLAND, N.Z.) 2022; 13:53-66. [PMID: 36091646 PMCID: PMC9462517 DOI: 10.2147/lctt.s335117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 08/23/2022] [Indexed: 06/15/2023]
Abstract
Arginine deprivation has gained increasing traction as a novel and safe antimetabolite strategy for the treatment of several hard-to-treat cancers characterised by a critical dependency on arginine. Small cell lung cancer (SCLC) displays marked arginine auxotrophy due to inactivation of the rate-limiting enzyme argininosuccinate synthetase 1 (ASS1), and as a consequence may be targeted with pegylated arginine deiminase or ADI-PEG20 (pegargiminase) and human recombinant pegylated arginases (rhArgPEG, BCT-100 and pegzilarginase). Although preclinical studies reveal that ASS1-deficient SCLC cell lines are highly sensitive to arginine-degrading enzymes, there is a clear disconnect with the clinic with minimal activity seen to date that may be due in part to patient selection. Recent studies have explored resistance mechanisms to arginine depletion focusing on tumor adaptation, such as ASS1 re-expression and autophagy, stromal cell inputs including macrophage infiltration, and tumor heterogeneity. Here, we explore how arginine deprivation may be combined strategically with novel agents to improve SCLC management by modulating resistance and increasing the efficacy of existing agents. Moreover, recent work has identified an intriguing role for targeting arginine in combination with PD-1/PD-L1 immune checkpoint inhibitors and clinical trials are in progress. Thus, future studies of arginine-depleting agents with chemoimmunotherapy, the current standard of care for SCLC, may lead to enhanced disease control and much needed improvements in long-term survival for patients.
Collapse
Affiliation(s)
- Joséphine Carpentier
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Iuliia Pavlyk
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Uma Mukherjee
- Department of Medical Oncology, Barts Health NHS Trust, St. Bartholomew’s Hospital, London, EC1A 7BE, UK
| | - Peter E Hall
- Department of Medical Oncology, Barts Health NHS Trust, St. Bartholomew’s Hospital, London, EC1A 7BE, UK
| | - Peter W Szlosarek
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
- Department of Medical Oncology, Barts Health NHS Trust, St. Bartholomew’s Hospital, London, EC1A 7BE, UK
| |
Collapse
|
6
|
Liu N, Shi F, Yang L, Liao W, Cao Y. Oncogenic viral infection and amino acid metabolism in cancer progression: Molecular insights and clinical implications. Biochim Biophys Acta Rev Cancer 2022; 1877:188724. [DOI: 10.1016/j.bbcan.2022.188724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 02/08/2023]
|
7
|
Coradduzza D, Solinas T, Azara E, Culeddu N, Cruciani S, Zinellu A, Medici S, Maioli M, Madonia M, Carru C. Plasma Polyamine Biomarker Panels: Agmatine in Support of Prostate Cancer Diagnosis. Biomolecules 2022; 12:biom12040514. [PMID: 35454104 PMCID: PMC9024899 DOI: 10.3390/biom12040514] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/10/2022] [Accepted: 03/26/2022] [Indexed: 01/03/2023] Open
Abstract
Prostate cancer is the most frequent malignant tumour among males (19%), often clinically silent and of difficult prognosis. Although several studies have highlighted the diagnostic and prognostic role of circulating biomarkers, such as PSA, their measurement does not necessarily allow the detection of the disease. Within this context, many authors suggest that the evaluation of circulating polyamines could represent a valuable tool, although several analytical problems still counteract their clinical practice. In particular, agmatine seems particularly intriguing, being a potential inhibitor of polyamines commonly derived from arginine. The aim of the present work was to evaluate the potential role of agmatine as a suitable biomarker for the identification of different classes of patients with prostate cancer (PC). For this reason, three groups of human patients—benign prostatic hyperplasia (BPH), precancerous lesion (PL), and prostate cancer (PC)—were recruited from a cohort of patients with suspected prostate cancer (n = 170), and obtained plasma was tested using the LC-HRMS method. Statistics on the receiver operating characteristics curve (ROC), and multivariate analysis were used to examine the predictive value of markers for discrimination among the three patient groups. Statistical analysis models revealed good discrimination using polyamine levels to distinguish the three classes of patients. AUC above 0.8, sensitivity ranging from 67% to 89%, specificity ranging from 74% to 89% and accuracy from 73% to 86%, considering the validation set, were achieved. Agmatine plasma levels were measured in PC (39.9 ± 12.06 ng/mL), BPH (77.62 ± 15.05 ng/mL), and PL (53.31 ± 15.27 ng/mL) patients. ROC analysis of the agmatine panel showed an AUC of 0.959 and p ≤ 0.001. These results could represent a future tool able to discriminate patients belonging to the three different clinical groups.
Collapse
Affiliation(s)
- Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.C.); (S.C.); (A.Z.); (M.M.)
| | - Tatiana Solinas
- Department of Clinical and Experimental Medicine, Urologic Clinic, University of Sassari, 07100 Sassari, Italy; (T.S.); (M.M.)
| | - Emanuela Azara
- Institute of Biomolecular Chemistry, National Research Council, 07100 Sassari, Italy; (E.A.); (N.C.)
| | - Nicola Culeddu
- Institute of Biomolecular Chemistry, National Research Council, 07100 Sassari, Italy; (E.A.); (N.C.)
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.C.); (S.C.); (A.Z.); (M.M.)
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.C.); (S.C.); (A.Z.); (M.M.)
| | - Serenella Medici
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.C.); (S.C.); (A.Z.); (M.M.)
| | - Massimo Madonia
- Department of Clinical and Experimental Medicine, Urologic Clinic, University of Sassari, 07100 Sassari, Italy; (T.S.); (M.M.)
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.C.); (S.C.); (A.Z.); (M.M.)
- Department of Biomedical Sciences and University Hospital of Sassari (AOU), 07100 Sassari, Italy
- Correspondence:
| |
Collapse
|
8
|
Fan K, Liu Z, Gao M, Tu K, Xu Q, Zhang Y. Targeting Nutrient Dependency in Cancer Treatment. Front Oncol 2022; 12:820173. [PMID: 35178349 PMCID: PMC8846368 DOI: 10.3389/fonc.2022.820173] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumor. Growing evidence suggests metabolic changes that support oncogenic progression may cause selective vulnerabilities that can be exploited for cancer treatment. Increasing demands for certain nutrients under genetic determination or environmental challenge enhance dependency of tumor cells on specific nutrient, which could be therapeutically developed through targeting such nutrient dependency. Various nutrients including several amino acids and glucose have been found to induce dependency in genetic alteration- or context-dependent manners. In this review, we discuss the extensively studied nutrient dependency and the biological mechanisms behind such vulnerabilities. Besides, existing applications and strategies to target nutrient dependency in different cancer types, accompanied with remaining challenges to further exploit these metabolic vulnerabilities to improve cancer therapies, are reviewed.
Collapse
Affiliation(s)
- Kexin Fan
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Second Medical College, Karamay, China
| | - Min Gao
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
| | - Yilei Zhang
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
9
|
Zhang Y, Higgins CB, Van Tine BA, Bomalaski JS, DeBosch BJ. Pegylated arginine deiminase drives arginine turnover and systemic autophagy to dictate energy metabolism. Cell Rep Med 2022; 3:100498. [PMID: 35106510 PMCID: PMC8784773 DOI: 10.1016/j.xcrm.2021.100498] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/16/2021] [Accepted: 12/16/2021] [Indexed: 12/30/2022]
Abstract
Obesity is a multi-systemic disorder of energy balance. Despite intense investigation, the determinants of energy homeostasis remain incompletely understood, and efficacious treatments against obesity and its complications are lacking. Here, we demonstrate that conferred arginine iminohydrolysis by the bacterial virulence factor and arginine deiminase, arcA, promotes mammalian energy expenditure and insulin sensitivity and reverses dyslipidemia, hepatic steatosis, and inflammation in obese mice. Extending this, pharmacological arginine catabolism via pegylated arginine deiminase (ADI-PEG 20) recapitulates these metabolic effects in dietary and genetically obese models. These effects require hepatic and whole-body expression of the autophagy complex protein BECN1 and hepatocyte-specific FGF21 secretion. Single-cell ATAC sequencing further reveals BECN1-dependent hepatocyte chromatin accessibility changes in response to ADI-PEG 20. The data thus reveal an unexpected therapeutic utility for arginine catabolism in modulating energy metabolism by activating systemic autophagy, which is now exploitable through readily available pharmacotherapy.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cassandra B. Higgins
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian A. Van Tine
- Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63108, USA
- Division of Pediatric Hematology/Oncology, St. Louis Children’s Hospital, St. Louis, MO 63108, USA
- Siteman Cancer Center, St. Louis, MO 63108, USA
| | | | - Brian J. DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Giatromanolaki A, Harris AL, Koukourakis MI. The prognostic and therapeutic implications of distinct patterns of argininosuccinate synthase 1 (ASS1) and arginase-2 (ARG2) expression by cancer cells and tumor stroma in non-small-cell lung cancer. Cancer Metab 2021; 9:28. [PMID: 34344457 PMCID: PMC8336070 DOI: 10.1186/s40170-021-00264-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/21/2021] [Indexed: 11/10/2022] Open
Abstract
Background Arginine (Arg) is essential for cancer cell growth and also for the activation of T cells. Thus, therapies aiming to reduce Arg utilization by cancer may prove detrimental for the immune response. Methods We examined the expression of two major enzymes involved in arginine depletion and replenishment, namely arginase ARG2 and argininosuccinate synthase ASS1, respectively, in a series of 98 NSCLCs. Their association with immune infiltrates and the postoperative outcome were also studied. Results ARG2 was expressed mainly by cancer-associated fibroblasts (CAFs) (58/98 cases; 59.2%), while ASS1 by cancer cells (75/98 cases; 76.5%). ASS1 and ARG2 expression patterns were not related to hypoxia markers. Auxotrophy, implied by the lack of expression of ASS1 in cancer cells, was associated with high angiogenesis (p < 0.02). ASS1 expression by cancer cells was associated with a high density of iNOS-expressing tumor-infiltrating lymphocytes (iNOS+TILs). ARG2 expression by CAFs was inversely related to the TIL-density and linked with poorer prognosis (p = 0.02). Patients with ASS1 expression by cancer cells had a better prognosis especially when CAFs did not express ARG2 (p = 0.004). Conclusions ARG2 and ASS1 enzymes are extensively expressed in NSCLC stroma and cancer cells, respectively. Auxotrophic tumors have a poor prognosis, potentially by utilizing Arg, thus reducing Arg-dependent TIL anti-tumor activity. ASS1 expression in cancer cells would allow Arg fueling of iNOS+TILs and enhance anti-tumor immunity. However, upregulation of ARG2 in CAFs may divert Arg from TILs, allowing immune escape. Identification of these three distinct phenotypes may be useful in the individualization of Arg-targeting therapies and immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-021-00264-7.
Collapse
Affiliation(s)
- Alexandra Giatromanolaki
- Department of Pathology, University Hospital of Alexandroupolis, Democritus University of Thrace, PO BOX 12, 68100, Alexandroupolis, Greece.,Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, PO BOX 12, 68100, Alexandroupolis, Greece
| | - Adrian L Harris
- Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Michael I Koukourakis
- Department of Pathology, University Hospital of Alexandroupolis, Democritus University of Thrace, PO BOX 12, 68100, Alexandroupolis, Greece. .,Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, PO BOX 12, 68100, Alexandroupolis, Greece.
| |
Collapse
|
11
|
Wang Z, Xie Q, Zhou H, Zhang M, Shen J, Ju D. Amino Acid Degrading Enzymes and Autophagy in Cancer Therapy. Front Pharmacol 2021; 11:582587. [PMID: 33510635 PMCID: PMC7836011 DOI: 10.3389/fphar.2020.582587] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Recently, there has been renewed interest in metabolic therapy for cancer, particularly in amino acid deprivation by enzymes. L-asparaginase was approved for the treatment of acute lymphoblastic leukemia by the U.S. Food and Drug Administration. Arginine deiminase and recombinant human arginase have been developed into clinical trials as potential cancer therapeutic agents for the treatment of arginine-auxotrophic tumors. Moreover, other novel amino acid degrading enzymes, such as glutaminase, methionase, lysine oxidase, phenylalanine ammonia lyase, have been developed for the treatment of malignant cancers. One of the greatest obstacles faced by anticancer drugs is the development of drug resistance, which is reported to be associated with autophagy. Autophagy is an evolutionarily conserved catabolic process that is responsible for the degradation of dysfunctional proteins and organelles. There is a growing body of literature revealing that, in response to metabolism stress, autophagy could be induced by amino acid deprivation. The manipulation of autophagy in combination with amino acid degrading enzymes is actively being investigated as a potential therapeutic approach in preclinical studies. Importantly, shedding light on how autophagy fuels tumor metabolism during amino acid deprivation will enable more potential combinational therapeutic strategies. This study summarizes recent advances, discussing several potential anticancer enzymes, and highlighting the promising combined therapeutic strategy of amino acid degrading enzymes and autophagy modulators in tumors
Collapse
Affiliation(s)
- Ziyu Wang
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Qinghong Xie
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| | - Haifeng Zhou
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China
| | - Min Zhang
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| |
Collapse
|
12
|
Bala K, Husain I, Sharma A. Arginine deaminase from Pseudomonas aeruginosa PS2: purification, biochemical characterization and in-vitro evaluation of anticancer activity. 3 Biotech 2020; 10:226. [PMID: 32373418 DOI: 10.1007/s13205-020-02212-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2023] Open
Abstract
In the present study, arginine deaminase (ADI) was purified from Pseudomonas aeruginosa PS2 which showed relative molecular mass of 70 ± 3 kDa on native-PAGE and 36 ± 0.5 kDa on SDS-PAGE. Purified ADI exhibited optimum activity at pH 6.5 and temperature 40 ºC. Metal ions, K+ and Mg2+ had positive, while Mn2+, Cr2+, Co2+, Fe3+, Ni2+, Cu2+, Cd2+ and Hg2+ had negative effects on catalytic activity of ADI. Purified enzyme showed high substrate specificity towards natural substrate L-arginine and did not hydrolyse its structural analogues. In-vitro serum half-life of purified ADI was 40 h, whereas proteolytic half-life was 28, 27, and 32 min against trypsin, elastase-I and proteinase-K, respectively. Anticancer activity of ADI has been evaluated against panel of human cancer cell lines (LS-180, HCT-116, MCF-7, BT-549, T47D, HL-60, MOLT-4, K-562, and PC-3) but lowest IC50 1.2 IU ml-1 was recorded with MCF-7 cells. Colony forming assay, wound-healing migration assay, phase contrast microscopy, DAPI staining, cell cycle analysis and DNA laddering assay revealed that ADI treatment induced apoptotic cell death in dose dependent manner. Increased level of MMP loss, ROS generation and decreased level of SOD, CAT, GPx and GSH displayed ADI treatment induced mitochondrial dysfunctioning. Furthermore, purified ADI had no substantial toxicity against human normal cell lines and blood erythrocytes. These findings suggesting that purified ADI could be developed as an anticancer agent but more in depth studies are warranted.
Collapse
Affiliation(s)
- Kiran Bala
- 1Bacteriology Laboratory, Department of P.G. Studies and Research in Biological Science, Rani Durgavati University, Jabalpur, Madhya Pradesh India
| | - Islam Husain
- 1Bacteriology Laboratory, Department of P.G. Studies and Research in Biological Science, Rani Durgavati University, Jabalpur, Madhya Pradesh India
- 2Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh India
| | - Anjana Sharma
- 1Bacteriology Laboratory, Department of P.G. Studies and Research in Biological Science, Rani Durgavati University, Jabalpur, Madhya Pradesh India
| |
Collapse
|
13
|
Agnello G, Alters SE, Rowlinson SW. Preclinical safety and antitumor activity of the arginine-degrading therapeutic enzyme pegzilarginase, a PEGylated, cobalt-substituted recombinant human arginase 1. Transl Res 2020; 217:11-22. [PMID: 31954097 DOI: 10.1016/j.trsl.2019.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 11/24/2022]
Abstract
Metabolic remodeling contributes to the development and progression of some cancers and exposes them to vulnerabilities, including specific nutrient dependencies that can be targeted therapeutically. Arginine is a semiessential amino acid, and several cancers are unable to endogenously synthesize sufficient levels of arginine for survival and proliferation, most commonly due to reduced expression of argininosuccinate synthase (ASS1). Such cancers are dependent on arginine and they can be targeted via enzyme-mediated depletion of systemic arginine. We report the preclinical safety, antitumor efficacy, and immune-potentiating effects of pegzilarginase, a highly potent human arginine-degrading enzyme. Toxicology studies showed that pegzilarginase-mediated arginine depletion is well tolerated at therapeutic levels that elicit an antitumor growth effect. To determine which tumor types are best suited for clinical development, we profiled clinical tumor samples for ASS1 expression, which correlated with pegzilarginase sensitivity in vivo in patient-derived xenograft (PDx) models. Among the histologies tested, malignant melanoma, small cell lung cancer and Merkel cell carcinoma had the highest prevalence of low ASS1 expression, the highest proportion of PDx models responding to pegzilarginase, and the strongest correlation between low or no ASS1 expression and sensitivity to pegzilarginase. In an immune-competent syngeneic mouse model, pegzilarginase slowed tumor growth and promoted the recruitment of CD8+ tumor infiltrating lymphocytes. This is consistent with the known autophagy-inducing effects of arginine depletion, and the link between autophagy and major histocompatibility complex antigen presentation to T cells. Our work supports the ongoing clinical investigations of pegzilarginase in solid tumors and clinical combination of pegzilarginase with immune checkpoint inhibitors.
Collapse
|
14
|
Harnessing cancer immunotherapy during the unexploited immediate perioperative period. Nat Rev Clin Oncol 2020; 17:313-326. [PMID: 32066936 DOI: 10.1038/s41571-019-0319-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
The immediate perioperative period (days before and after surgery) is hypothesized to be crucial in determining long-term cancer outcomes: during this short period, numerous factors, including excess stress and inflammatory responses, tumour-cell shedding and pro-angiogenic and/or growth factors, might facilitate the progression of pre-existing micrometastases and the initiation of new metastases, while simultaneously jeopardizing immune control over residual malignant cells. Thus, application of anticancer immunotherapy during this critical time frame could potentially improve patient outcomes. Nevertheless, this strategy has rarely been implemented to date. In this Perspective, we discuss apparent contraindications for the perioperative use of cancer immunotherapy, suggest safe immunotherapeutic and other anti-metastatic approaches during this important time frame and specify desired characteristics of such interventions. These characteristics include a rapid onset of immune activation, avoidance of tumour-promoting effects, no or minimal increase in surgical risk, resilience to stress-related factors and minimal induction of stress responses. Pharmacological control of excess perioperative stress-inflammatory responses has been shown to be clinically feasible and could potentially be combined with immune stimulation to overcome the direct pro-metastatic effects of surgery, prevent immune suppression and enhance immunostimulatory responses. Accordingly, we believe that certain types of immunotherapy, together with interventions to abrogate stress-inflammatory responses, should be evaluated in conjunction with surgery and, for maximal effectiveness, could be initiated before administration of adjuvant therapies. Such strategies might improve the overall success of cancer treatment.
Collapse
|
15
|
Zarei M, Rahbar MR, Negahdaripour M, Morowvat MH, Nezafat N, Ghasemi Y. Cell Penetrating Peptide: Sequence-Based Computational Prediction for Intercellular Delivery of Arginine Deiminase. CURR PROTEOMICS 2020. [DOI: 10.2174/1570164616666190701120351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Cell-Penetrating Peptides (CPPs), a family of short peptides, are broadly used as the carrier in the delivery of drugs and different therapeutic agents. Thanks to the existence of valuable databases, computational screening of the experimentally validated CPPs can help the researchers to select more effective CPPs for the intercellular delivery of therapeutic proteins. Arginine deiminase of Mycoplasma hominis, an arginine-degrading enzyme, is currently in the clinical trial for treating several arginine auxotrophic cancers. However, some tumor cells have developed resistance to ADI treatment. The ADI resistance arises from the over-expression of argininosuccinate synthetase 1 enzyme, which is involved in arginine synthesis. Intracellular delivery of ADI into tumor cells is suggested as an efficient approach to overcome the aforesaid drawback.Objective:In this study, in-silico tools were used for evaluating the experimentally validated CPPs to select the best CPP candidates for the intracellular delivery of ADI.Results:In this regard, 150 CPPs of protein cargo available at CPPsite were retrieved and evaluated by the CellPPD server. The best CPP candidates for the intracellular delivery of ADI were selected based on stability and antigenicity of the ADI-CPP fusion form. The conjugated forms of ADI with each of the three CPPs including EGFP-hcT (9-32), EGFP-ppTG20, and F(SG)4TP10 were stable and nonantigenic; thus, these sequences were introduced as the best CPP candidates for the intracellular delivery of ADI. In addition, the proposed CPPs had appropriate positive charge and lengths for an efficient cellular uptake.Conclusion:These three introduced CPPs not only are appropriate for the intracellular delivery of ADI, but also can overcome the limitation of its therapeutic application, including short half-life and antigenicity.
Collapse
Affiliation(s)
- Mahboubeh Zarei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Pokrovsky VS, Chepikova OE, Davydov DZ, Zamyatnin AA, Lukashev AN, Lukasheva EV. Amino Acid Degrading Enzymes and their Application in Cancer Therapy. Curr Med Chem 2019; 26:446-464. [PMID: 28990519 DOI: 10.2174/0929867324666171006132729] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 09/12/2017] [Accepted: 09/28/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Amino acids are essential components in various biochemical pathways. The deprivation of certain amino acids is an antimetabolite strategy for the treatment of amino acid-dependent cancers which exploits the compromised metabolism of malignant cells. Several studies have focused on the development and preclinical and clinical evaluation of amino acid degrading enzymes, namely L-asparaginase, L-methionine γ-lyase, L-arginine deiminase, L-lysine α-oxidase. Further research into cancer cell metabolism may therefore define possible targets for controlling tumor growth. OBJECTIVE The purpose of this review was to summarize recent progress in the relationship between amino acids metabolism and cancer therapy, with a particular focus on Lasparagine, L-methionine, L-arginine and L-lysine degrading enzymes and their formulations, which have been successfully used in the treatment of several types of cancer. METHODS We carried out a structured search among literature regarding to amino acid degrading enzymes. The main aspects of search were in vitro and in vivo studies, clinical trials concerning application of these enzymes in oncology. RESULTS Most published research are on the subject of L-asparaginase properties and it's use for cancer treatment. L-arginine deiminase has shown promising results in a phase II trial in advanced melanoma and hepatocellular carcinoma. Other enzymes, in particular Lmethionine γ-lyase and L-lysine α-oxidase, were effective in vitro and in vivo. CONCLUSION The findings of this review revealed that therapy based on amino acid depletion may have the potential application for cancer treatment but further clinical investigations are required to provide the efficacy and safety of these agents.
Collapse
Affiliation(s)
- Vadim S Pokrovsky
- Blokhin Cancer Research Center, Moscow, Russian Federation.,Orekhovich Institute of Biomedical Chemistry, Moscow, Russian Federation.,People's Friendship University, Russia (RUDN University), Moscow, Russian Federation
| | - Olga E Chepikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Belozersky Institute of Physico- Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Alexander N Lukashev
- People's Friendship University, Russia (RUDN University), Moscow, Russian Federation.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Elena V Lukasheva
- People's Friendship University, Russia (RUDN University), Moscow, Russian Federation
| |
Collapse
|
17
|
Zarei M, Rahbar MR, Morowvat MH, Nezafat N, Negahdaripour M, Berenjian A, Ghasemi Y. Arginine Deiminase: Current Understanding and Applications. Recent Pat Biotechnol 2019; 13:124-136. [PMID: 30569861 DOI: 10.2174/1872208313666181220121400] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/07/2018] [Accepted: 12/25/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Arginine deiminase (ADI), an arginine catabolizing enzyme, is considered as an anti-tumor agent for the treatment of arginine auxotrophic cancers. However, some obstacles limit its clinical applications. OBJECTIVE This review will summarize the clinical applications of ADI, from a brief history to its limitations, and will discuss the different ways to deal with the clinical limitations. METHOD The structure analysis, cloning, expression, protein engineering and applications of arginine deiminase enzyme have been explained in this review. CONCLUSION Recent patents on ADI are related to ADI engineering to increase its efficacy for clinical application. The intracellular delivery of ADI and combination therapy seem to be the future strategies in the treatment of arginine auxotrophic cancers. Applying ADIs with optimum features from different sources and or ADI engineering, are promising strategies to improve the clinical application of ADI.
Collapse
Affiliation(s)
- Mahboubeh Zarei
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Rahbar
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Morowvat
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aydin Berenjian
- School of Engineering, Faculty of Science & Engineering, The University of Waikato, Hamilton, New Zealand
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Yeon A, You S, Kim M, Gupta A, Park MH, Weisenberger DJ, Liang G, Kim J. Rewiring of cisplatin-resistant bladder cancer cells through epigenetic regulation of genes involved in amino acid metabolism. Theranostics 2018; 8:4520-4534. [PMID: 30214636 PMCID: PMC6134931 DOI: 10.7150/thno.25130] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/23/2018] [Indexed: 12/26/2022] Open
Abstract
Alterations in DNA methylation are important epigenetic markers in bladder cancer (BC). These epigenome modifications may drive the mechanisms of aggressive chemo-resistant BC. Clinicopathological biomarkers that indicate chemotherapeutic resistance are critical for better assessing treatment strategies for individual patients. Thus, in this study, we aimed to determine whether DNA methylation of certain metabolic enzymes is significantly altered in cisplatin-resistant BC cells. Methods: To characterize CpG methylation and nucleosome accessibility in cisplatin-resistant BC cells, the Illumina Infinium HM450 DNA methylation assay was performed. Perturbed gene expression was found to be associated with cisplatin resistance, and the biological roles of spermidine/spermine N1-acetyltransferase (SAT1) and argininosuccinate synthase 1 (ASS1) were further studied using qRT-PCR analysis and various cell biology assays, including western blot. Results:ASS1 and SAT1, genes for amino acid and polyamine metabolism catalysts, respectively, were found to be vastly hypermethylated, resulting in greatly downregulated expression. ASS1 expression is of particular interest because prior studies have demonstrated its potential association with BC stage and recurrence. In regard to chemoresistance, we found that aberrant expression or induced stimulation of SAT1 restored cisplatin sensitivity in the cell culture system. We also found that the addition of exogenous arginine deiminase through administration of ADI-PEG 20 (pegylated arginine deiminase) increased ASS1 expression and enhanced cisplatin's apoptotic effects. Conclusions: Our study demonstrates a novel mechanistic link between the epigenetic perturbation of SAT1 and ASS1 and cancer metabolism in cisplatin-resistant bladder cancer cells. These findings suggest potential utility of SAT1 and ASS1 as predictive biomarkers in re-sensitizing bladder cancer to chemotherapy and personalizing therapy.
Collapse
Affiliation(s)
- Austin Yeon
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Minhyung Kim
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Amit Gupta
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Myung Hee Park
- National Institute of Dental and Craniofacial Research, National Institutes of Health Bethesda, MD, USA
| | - Daniel J. Weisenberger
- Department of Biochemistry and Molecular Medicine, USC Norris Comprehensive Cancer Center, University of Southern California
| | - Gangning Liang
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jayoung Kim
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Departments of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, University of California Los Angeles, CA, USA
- Department of Urology, Ga Cheon University College of Medicine, Incheon, Republic of Korea
| |
Collapse
|
19
|
Jiang H, Guo S, Xiao D, Bian X, Wang J, Wang Y, Zhou H, Cai J, Zheng Z. Arginine deiminase expressed in vivo, driven by human telomerase reverse transcriptase promoter, displays high hepatoma targeting and oncolytic efficiency. Oncotarget 2018; 8:37694-37704. [PMID: 28455966 PMCID: PMC5514941 DOI: 10.18632/oncotarget.17032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/21/2017] [Indexed: 12/23/2022] Open
Abstract
Arginine starvation has the potential to selectively treat both primary tumor and (micro) metastatic tissue with very low side effects. Arginine deiminase (ADI; EC 3.5.3.6), an arginine-degrading enzyme, has been studied as a potential anti-tumor drug for the treatment of arginine-auxotrophic tumors. Though ADI-PEG20 (pegylated ADI by PEG 20,000) already passed the phase I/II clinical trials [1], it is just used as adjuvant therapy because of its low efficiency and less targeting. Then, this paper discussed the efficiency of arginine starvation mediated by ADI expressed in cytoplasm for liver cancers. In order to guarantee the tumor targeting, human telomerase reverse transcriptase (hTERT) promoter was used to drive the expression of ADI in vivo. To access the anti-tumor efficiency of ADI, p53 gene was used as the positive control. Thus, ADI displayed obvious cytotoxicity to BEL7402 and HUH7 cell lines in cytoplasm. The apoptosis rates rose from 15% to nearly 60% after changing the expression vectors from pcDNA4 plasmid to adenovirus. Compared with p53-adenovirus, ADI-adenovirus showed the higher oncolytic activity in the intratumoral injection model of mice. Tumor disappeared after the treatment of ADI-adenovirus for two weeks, and the mice pulled through all. Therefore, ADI is an ideal anti-tumor gene for caner targeting therapy with the help of hTERT promoter.
Collapse
Affiliation(s)
- Hui Jiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Song Guo
- Department of Orthopedic, Wuhan Puai Hospital, Wuhan 430034, China
| | - Dan Xiao
- Department of Gastroenterology, Jianghan University Affiliated Hospital, Wuhan 430000, China
| | - Xuzhao Bian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jie Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ying Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huiting Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jun Cai
- Hubei Collaborative Innovation Center for Industrial Fermentation, College of Biotechnology, Hubei University of Technology, Wuhan 430068, China
| | - Zhongliang Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
20
|
Maletzki C, Rosche Y, Riess C, Scholz A, William D, Classen CF, Kreikemeyer B, Linnebacher M, Fiedler T. Deciphering molecular mechanisms of arginine deiminase-based therapy - Comparative response analysis in paired human primary and recurrent glioblastomas. Chem Biol Interact 2017; 278:179-188. [PMID: 28989041 DOI: 10.1016/j.cbi.2017.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 12/23/2022]
Abstract
Arginine auxotrophy constitutes the Achilles' heel for several tumors, among them glioblastoma multiforme (GBM). Hence, arginine-depleting enzymes such as arginine deiminase (ADI) from Streptococcus pyogenes are promising for treatment of primary and maybe even refractory GBM. Based on our previous study in which ADI-susceptibility was shown on a panel of patient-derived GBM cell lines, we here aimed at deciphering underlying molecular mechanisms of ADI-mediated growth inhibition. We found that ADI (35 mU/mL) initially induces a cellular stress-response that is characterized by upregulation of genes primarily belonging to the heat-shock protein family. In addition to autophagocytosis, we show for the first time that senescence constitutes another cellular response mechanism upon ADI-treatment and that this bacterial enzyme is able to act as radiosensitizer (¼ cases). Long-term treatment schedules revealed no resistance development, with treated cells showing morphological signs of cell stress. Next, several combination strategies were employed to optimize ADI-based treatment. Simultaneous and sequential S. pyogenes ADI-based combinations included substances acting at different molecular pathways (curcumin, resveratrol, quinacrine, and sorafenib, 2 × 72 h treatment). Adding drugs to GBM cell lines (n = 4, including a matched pair of primary and recurrent GBM in one case) accelerated and potentiated ADI-mediated cytotoxicity. Autophagy was identified as the main cause of tumor growth inhibition. Of note, residual cells again showed classical signs of senescence in most combinations. Our results suggest an alternative treatment regimen for this fatal cancer type which circumvents many of the traditional barriers. Using the metabolic defect in GBM thus warrants further (pre-) clinical evaluation.
Collapse
Affiliation(s)
- Claudia Maletzki
- Molecular Oncology and Immunotherapy, Department of General Surgery, 18057 Rostock, Germany.
| | - Yvonne Rosche
- Molecular Oncology and Immunotherapy, Department of General Surgery, 18057 Rostock, Germany; Institute for Medical Microbiology, Virology, and Hygiene, 18057 Rostock, Germany
| | - Christin Riess
- Molecular Oncology and Immunotherapy, Department of General Surgery, 18057 Rostock, Germany; Institute for Medical Microbiology, Virology, and Hygiene, 18057 Rostock, Germany
| | - Aline Scholz
- Molecular Oncology and Immunotherapy, Department of General Surgery, 18057 Rostock, Germany; Institute for Medical Microbiology, Virology, and Hygiene, 18057 Rostock, Germany
| | - Doreen William
- Molecular Oncology and Immunotherapy, Department of General Surgery, 18057 Rostock, Germany; University Childrens' Hospital, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Carl Friedrich Classen
- University Childrens' Hospital, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Bernd Kreikemeyer
- Institute for Medical Microbiology, Virology, and Hygiene, 18057 Rostock, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Department of General Surgery, 18057 Rostock, Germany
| | - Tomas Fiedler
- Institute for Medical Microbiology, Virology, and Hygiene, 18057 Rostock, Germany
| |
Collapse
|
21
|
Sharma A, Bala K, Husain I. Preliminary evaluation of arginine deiminase activity of indigenous bacterial strains for suitable chemotherapeutic applications. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2017. [DOI: 10.1016/j.bcab.2017.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Fung MKL, Chan GCF. Drug-induced amino acid deprivation as strategy for cancer therapy. J Hematol Oncol 2017; 10:144. [PMID: 28750681 PMCID: PMC5530962 DOI: 10.1186/s13045-017-0509-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/10/2017] [Indexed: 12/21/2022] Open
Abstract
Cancer is caused by uncontrollable growth of neoplastic cells, leading to invasion of adjacent and distant tissues resulting in death. Cancer cells have specific nutrient(s) auxotrophy and have a much higher nutrient demand compared to normal tissues. Therefore, different metabolic inhibitors or nutrient-depleting enzymes have been tested for their anti-cancer activities. We review recent available laboratory and clinical data on using various specific amino acid metabolic pathways inhibitors in treating cancers. Our focus is on glutamine, asparagine, and arginine starvation. These three amino acids are chosen due to their better scientific evidence compared to other related approaches in cancer treatment. Amino acid-specific depleting enzymes have been adopted in different standard chemotherapy protocols. Glutamine starvation by glutaminase inhibitior, transporter inhibitor, or glutamine depletion has shown to have significant anti-cancer effect in pre-clinical studies. Currently, glutaminase inhibitor is under clinical trial for testing anti-cancer efficacy. Clinical data suggests that asparagine depletion is effective in treating hematologic malignancies even as a single agent. On the other hand, arginine depletion has lower toxicity profile and can effectively reduce the level of pro-cancer biochemicals in patients as shown by ours and others’ data. This supports the clinical use of arginine depletion as anti-cancer therapy but its exact efficacy in various cancers requires further investigation. However, clinical application of these enzymes is usually hindered by common problems including allergy to these foreign proteins, off-target cytotoxicity, short half-life and rapidly emerging chemoresistance. There have been efforts to overcome these problems by modifying the drugs in different ways to circumvent these hindrance such as (1) isolate human native enzymes to reduce allergy, (2) isolate enzyme isoforms with higher specificities and efficiencies, (3) pegylate the enzymes to reduce allergy and prolong the half-lives, and (4) design drug combinations protocols to enhance the efficacy of chemotherapy by drug synergy and minimizing resistance. These improvements can potentially lead to the development of more effective anti-cancer treatment with less adverse effects and higher therapeutic efficacy.
Collapse
Affiliation(s)
- Marcus Kwong Lam Fung
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
23
|
Miyamoto T, Lo PHY, Saichi N, Ueda K, Hirata M, Tanikawa C, Matsuda K. Argininosuccinate synthase 1 is an intrinsic Akt repressor transactivated by p53. SCIENCE ADVANCES 2017; 3:e1603204. [PMID: 28560349 PMCID: PMC5438217 DOI: 10.1126/sciadv.1603204] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/22/2017] [Indexed: 06/07/2023]
Abstract
The transcription factor p53 is at the core of a built-in tumor suppression system that responds to varying degrees of stress input and is deregulated in most human cancers. Befitting its role in maintaining cellular fitness and fidelity, p53 regulates an appropriate set of target genes in response to cellular stresses. However, a comprehensive understanding of this scheme has not been accomplished. We show that argininosuccinate synthase 1 (ASS1), a citrulline-aspartate ligase in de novo arginine synthesis pathway, was directly transactivated by p53 in response to genotoxic stress, resulting in the rearrangement of arginine metabolism. Furthermore, we found that x-ray irradiation promoted the systemic induction of Ass1 and concomitantly increased plasma arginine levels in p53+/+ mice but not in p53-/- mice. Notably, Ass1+/- mice exhibited hypersensitivity to whole-body irradiation owing to increased apoptosis in the small intestinal crypts. Analyses of ASS1-deficient cells generated using the CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated 9) system revealed that ASS1 plays a pivotal role in limiting Akt phosphorylation. In addition, aberrant activation of Akt resulting from ASS1 loss disrupted Akt-mediated cell survival signaling activity under genotoxic stress. Building on these results, we demonstrated that p53 induced an intrinsic Akt repressor, ASS1, and the perturbation of ASS1 expression rendered cells susceptible to genotoxic stress. Our findings uncover a new function of p53 in the regulation of Akt signaling and reveal how p53, ASS1, and Akt are interrelated to each other.
Collapse
Affiliation(s)
- Takafumi Miyamoto
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Paulisally Hau Yi Lo
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Naomi Saichi
- Cancer Proteomics Group, Genome Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Genome Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Hirata
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Chizu Tanikawa
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
Zauderer MG. Standard Chemotherapy Options and Clinical Trials of Novel Agents for Mesothelioma. ASBESTOS AND MESOTHELIOMA 2017. [DOI: 10.1007/978-3-319-53560-9_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
25
|
McNeal CJ, Meininger CJ, Reddy D, Wilborn CD, Wu G. Safety and Effectiveness of Arginine in Adults. J Nutr 2016; 146:2587S-2593S. [PMID: 27934649 DOI: 10.3945/jn.116.234740] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/05/2016] [Accepted: 09/21/2016] [Indexed: 02/06/2023] Open
Abstract
l-Arginine (Arg) appears to have a beneficial effect on the regulation of nutrient metabolism to enhance lean tissue deposition and on insulin resistance in humans. The observed safe level for oral administration of Arg is ∼20 g/d, but higher levels have been tested in short-term studies without serious adverse effects; however, more data are needed in both animal models and humans to fully evaluate safety as well as efficacy. The primary objective of this review is to summarize the current knowledge of the safety, pharmacokinetics, and effectiveness of oral Arg in adults. Arg supplementation has been used safely in vulnerable populations, such as pregnant women, preterm infants, and individuals with cystic fibrosis. Several recent studies have shown beneficial effects of Arg in individuals with obesity, insulin resistance, and diabetes. Collectively, the data suggest that Arg supplementation is a safe and generally well-tolerated nutriceutical that may improve metabolic profiles in humans.
Collapse
Affiliation(s)
- Catherine J McNeal
- Department of Internal Medicine, Baylor Scott & White Health, Temple, TX;
| | - Cynthia J Meininger
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Temple, TX
| | - Deepika Reddy
- University Diabetes and Endocrinology Center, University of Utah Health Science Center, Salt Lake City, UT
| | - Colin D Wilborn
- Department of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX; and
| | - Guoyao Wu
- Department of Animal Science and Faculty of Nutrition, Texas A&M University, College Station, TX
| |
Collapse
|
26
|
Locke M, Ghazaly E, Freitas MO, Mitsinga M, Lattanzio L, Lo Nigro C, Nagano A, Wang J, Chelala C, Szlosarek P, Martin SA. Inhibition of the Polyamine Synthesis Pathway Is Synthetically Lethal with Loss of Argininosuccinate Synthase 1. Cell Rep 2016; 16:1604-1613. [PMID: 27452468 PMCID: PMC4978703 DOI: 10.1016/j.celrep.2016.06.097] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/09/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022] Open
Abstract
Argininosuccinate synthase 1 (ASS1) is the rate-limiting enzyme for arginine biosynthesis. ASS1 expression is lost in a range of tumor types, including 50% of malignant pleural mesotheliomas. Starving ASS1-deficient cells of arginine with arginine blockers such as ADI-PEG20 can induce selective lethality and has shown great promise in the clinical setting. We have generated a model of ADI-PEG20 resistance in mesothelioma cells. This resistance is mediated through re-expression of ASS1 via demethylation of the ASS1 promoter. Through coordinated transcriptomic and metabolomic profiling, we have shown that ASS1-deficient cells have decreased levels of acetylated polyamine metabolites, together with a compensatory increase in the expression of polyamine biosynthetic enzymes. Upon arginine deprivation, polyamine metabolites are decreased in the ASS1-deficient cells and in plasma isolated from ASS1-deficient mesothelioma patients. We identify a synthetic lethal dependence between ASS1 deficiency and polyamine metabolism, which could potentially be exploited for the treatment of ASS1-negative cancers.
Collapse
Affiliation(s)
- Matthew Locke
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Essam Ghazaly
- Centre for Haemato-oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Marta O Freitas
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Mikaella Mitsinga
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Laura Lattanzio
- Laboratorio di Genetica Oncologica ed Oncologia Translazionale and Dipartimento di Oncologia, Azienda Ospedaliera S. Croce e Carle, 12100 Cuneo, Italy
| | - Cristiana Lo Nigro
- Laboratorio di Genetica Oncologica ed Oncologia Translazionale and Dipartimento di Oncologia, Azienda Ospedaliera S. Croce e Carle, 12100 Cuneo, Italy
| | - Ai Nagano
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jun Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Peter Szlosarek
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sarah A Martin
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
27
|
Affiliation(s)
- Fabricio Loayza-Puch
- a Division of Biological Stress Response, The Netherlands Cancer Institute , Amsterdam , The Netherlands
| | - Reuven Agami
- a Division of Biological Stress Response, The Netherlands Cancer Institute , Amsterdam , The Netherlands.,b Erasmus MC, Rotterdam University , Rotterdam , The Netherlands
| |
Collapse
|
28
|
Patil MD, Bhaumik J, Babykutty S, Banerjee UC, Fukumura D. Arginine dependence of tumor cells: targeting a chink in cancer's armor. Oncogene 2016; 35:4957-72. [PMID: 27109103 DOI: 10.1038/onc.2016.37] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/02/2016] [Accepted: 02/02/2016] [Indexed: 12/14/2022]
Abstract
Arginine, one among the 20 most common natural amino acids, has a pivotal role in cellular physiology as it is being involved in numerous cellular metabolic and signaling pathways. Dependence on arginine is diverse for both tumor and normal cells. Because of decreased expression of argininosuccinate synthetase and/or ornithine transcarbamoylase, several types of tumor are auxotrophic for arginine. Deprivation of arginine exploits a significant vulnerability of these tumor cells and leads to their rapid demise. Hence, enzyme-mediated arginine depletion is a potential strategy for the selective destruction of tumor cells. Arginase, arginine deiminase and arginine decarboxylase are potential enzymes that may be used for arginine deprivation therapy. These arginine catabolizing enzymes not only reduce tumor growth but also make them susceptible to concomitantly administered anti-cancer therapeutics. Most of these enzymes are currently under clinical investigations and if successful will potentially be advanced as anti-cancer modalities.
Collapse
Affiliation(s)
- M D Patil
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Punjab, India
| | - J Bhaumik
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Punjab, India
| | - S Babykutty
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - U C Banerjee
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Punjab, India
| | - D Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Patil MD, Shinde KD, Patel G, Chisti Y, Banerjee UC. Use of response surface method for maximizing the production of arginine deiminase by Pseudomonas putida. ACTA ACUST UNITED AC 2016; 10:29-37. [PMID: 28352521 PMCID: PMC5070923 DOI: 10.1016/j.btre.2016.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/23/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
First report on Arginine deiminase production from
Pseudomonas putida using RSM. Optimum conditions for ADI production were established in
shake flasks. ADI production was assessed in a 14 L
bioreactor under the optimized conditions. Repressor effect of aeration on ADI production was
observed in 14 L bioreactor. Substantial improvement of 4.5-folds in ADI titre was
achieved.
Statistically designed experiments were used to optimize
the production of arginine deiminase (ADI) by Pseudomonas
putida KT2440 in batch culture. A Plackett-Burman design involving
eleven factors showed that ADI production was most influenced by the initial pH and
the initial concentrations of glucose and yeast extract. A central composite
experimental design showed that the optimal values of these factors were 8.0,
10 g/L and 12.5 g/L, respectively. The other
components of the optimal culture medium were bacto peptone 7.5 g/L, Triton X–100 0.30% (v/v), and arginine 3 g/L, for a culture
temperature of 25 °C. Compared with the basal medium, the ADI
activity in the optimized medium had nearly 4.5-fold increase (4.31 U/mL). The optimized medium was then used for a further study of ADI production in
a 14 L stirred tank bioreactor. The agitation speed and the
aeration rates were varied to determine suitable values of these
variables.
Collapse
Affiliation(s)
- Mahesh D Patil
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, 160062 Punjab, India
| | - Kiran D Shinde
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, 160062 Punjab, India
| | - Gopal Patel
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, 160062 Punjab, India
| | - Yusuf Chisti
- School of Engineering, Massey University, Private Bag 11 222, Palmerston North, New Zealand
| | - Uttam Chand Banerjee
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, 160062 Punjab, India
| |
Collapse
|
30
|
Abstract
In this issue of Blood, Miraki-Moud et al demonstrate that the majority of acute myeloid leukemias (AMLs) have low expression of argininosuccinate synthetase-1 (ASS1), rendering AML cell lines and primary AML blasts dependent on exogenous arginine and sensitized to arginine deprivation.
Collapse
|
31
|
Qiu F, Huang J, Sui M. Targeting arginine metabolism pathway to treat arginine-dependent cancers. Cancer Lett 2015; 364:1-7. [DOI: 10.1016/j.canlet.2015.04.020] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/28/2015] [Accepted: 04/19/2015] [Indexed: 01/01/2023]
|
32
|
Mussai F, Egan S, Higginbotham-Jones J, Perry T, Beggs A, Odintsova E, Loke J, Pratt G, U KP, Lo A, Ng M, Kearns P, Cheng P, De Santo C. Arginine dependence of acute myeloid leukemia blast proliferation: a novel therapeutic target. Blood 2015; 125:2386-96. [PMID: 25710880 PMCID: PMC4416943 DOI: 10.1182/blood-2014-09-600643] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common acute leukemias in adults and children, yet significant numbers of patients relapse and die of disease. In this study, we identify the dependence of AML blasts on arginine for proliferation. We show that AML blasts constitutively express the arginine transporters CAT-1 and CAT-2B, and that the majority of newly diagnosed patients' blasts have deficiencies in the arginine-recycling pathway enzymes argininosuccinate synthase and ornithine transcarbamylase, making them arginine auxotrophic. BCT-100, a pegylated human recombinant arginase, leads to a rapid depletion in extracellular and intracellular arginine concentrations, resulting in arrest of AML blast proliferation and a reduction in AML engraftment in vivo. BCT-100 as a single agent causes significant death of AML blasts from adults and children, and acts synergistically in combination with cytarabine. Using RNA sequencing, 20 further candidate genes which correlated with resistance have been identified. Thus, AML blasts are dependent on arginine for survival and proliferation, as well as depletion of arginine with BCT-100 of clinical value in the treatment of AML.
Collapse
MESH Headings
- Adolescent
- Aged
- Animals
- Antimetabolites, Antineoplastic/therapeutic use
- Arginase/therapeutic use
- Arginine/metabolism
- Child
- Child, Preschool
- Cytarabine/therapeutic use
- Enzyme Therapy
- Female
- Humans
- Infant
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice, SCID
- Middle Aged
- Recombinant Proteins/therapeutic use
- Tumor Cells, Cultured
- Young Adult
Collapse
Affiliation(s)
- Francis Mussai
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sharon Egan
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | | | - Tracey Perry
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Andrew Beggs
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Elena Odintsova
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Justin Loke
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Guy Pratt
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Kin Pong U
- Bio-cancer Treatment International Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong; and
| | - Anthony Lo
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Margaret Ng
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Pamela Kearns
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Cheng
- Bio-cancer Treatment International Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong; and
| | - Carmela De Santo
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
33
|
Fiedler T, Strauss M, Hering S, Redanz U, William D, Rosche Y, Classen CF, Kreikemeyer B, Linnebacher M, Maletzki C. Arginine deprivation by arginine deiminase of Streptococcus pyogenes controls primary glioblastoma growth in vitro and in vivo. Cancer Biol Ther 2015; 16:1047-55. [PMID: 25774632 DOI: 10.1080/15384047.2015.1026478] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Arginine auxotrophy constitutes a weak point of several tumors, among them glioblastoma multiforme (GBM). Hence, those tumors are supposed to be sensitive for arginine-depleting substances, such as arginine deiminase (ADI). Here we elucidated the sensitivity of patient-individual GBM cell lines toward Streptococcus pyogenes-derived ADI. To improve therapy, ADI was combined with currently established and pre-clinical cytostatic drugs. Additionally, effectiveness of local ADI therapy was determined in xenopatients. Half of the GBM cell lines tested responded well toward ADI monotherapy. In those cell lines, viability decreased significantly (up to 50%). Responding cell lines were subjected to combination therapy experiments to test if any additive or even synergistic effects may be achieved. Such promising results were obtained in 2/3 cases. In cell lines HROG02, HROG05 and HROG10, ADI and Palomid 529 combinations were most effective yielding more than 70% killing after 2 rounds of treatment. Comparable boosted antitumoral effects were observed after adding chloroquine to ADI (>60% killing). Apoptosis, as well as cell cycle dysregulation were found to play a minor role. In some, but clearly not all cases, (epi-) genetic silencing of arginine synthesis pathway genes (argininosuccinate synthetase 1 and argininosuccinate lyase) explained obtained results. In vivo, ADI as well as the combination of ADI and SAHA efficiently controlled HROG05 xenograft growth, whereas adding Palomid 529 to ADI did not further increase the strong antitumoral effect of ADI. The cumulative in vitro and in vivo results proved ADI as a very promising candidate therapeutic, especially for development of adjuvant GBM combination treatments.
Collapse
Affiliation(s)
- Tomas Fiedler
- a Institute for Medical Microbiology, Virology, and Hygiene; Rostock University Medical Centre ; Rostock , Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Marini JC, Didelija IC. Arginine depletion by arginine deiminase does not affect whole protein metabolism or muscle fractional protein synthesis rate in mice. PLoS One 2015; 10:e0119801. [PMID: 25775142 PMCID: PMC4361593 DOI: 10.1371/journal.pone.0119801] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/20/2015] [Indexed: 01/23/2023] Open
Abstract
Due to the absolute need for arginine that certain cancer cells have, arginine depletion is a therapy in clinical trials to treat several types of cancers. Arginine is an amino acids utilized not only as a precursor for other important molecules, but also for protein synthesis. Because arginine depletion can potentially exacerbate the progressive loss of body weight, and especially lean body mass, in cancer patients we determined the effect of arginine depletion by pegylated arginine deiminase (ADI-PEG 20) on whole body protein synthesis and fractional protein synthesis rate in multiple tissues of mice. ADI-PEG 20 successfully depleted circulating arginine (<1 μmol/L), and increased citrulline concentration more than tenfold. Body weight and body composition, however, were not affected by ADI-PEG 20. Despite the depletion of arginine, whole body protein synthesis and breakdown were maintained in the ADI-PEG 20 treated mice. The fractional protein synthesis rate of muscle was also not affected by arginine depletion. Most tissues (liver, kidney, spleen, heart, lungs, stomach, small and large intestine, pancreas) were able to maintain their fractional protein synthesis rate; however, the fractional protein synthesis rate of brain, thymus and testicles was reduced due to the ADI-PEG 20 treatment. Furthermore, these results were confirmed by the incorporation of ureido [14C]citrulline, which indicate the local conversion into arginine, into protein. In conclusion, the intracellular recycling pathway of citrulline is able to provide enough arginine to maintain protein synthesis rate and prevent the loss of lean body mass and body weight.
Collapse
Affiliation(s)
- Juan C. Marini
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| | - Inka Cajo Didelija
- United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
35
|
Argininosuccinate synthetase (ASS) deficiency in high-grade pulmonary neuroendocrine carcinoma: an opportunity for personalized targeted therapy. J Cancer Res Clin Oncol 2014; 141:1363-9. [DOI: 10.1007/s00432-014-1904-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/21/2014] [Indexed: 11/25/2022]
|