1
|
Prasad YR, Anakha J, Jawalekar SS, Pande AH. Broad-spectrum anti-cancer activity of fused human arginase variants. Invest New Drugs 2024; 42:531-537. [PMID: 39160429 DOI: 10.1007/s10637-024-01466-8] [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: 07/15/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
The rapid increase in cancer cases worldwide necessitates the development of novel therapeutic approaches. Therapies targeting cancer's altered metabolism, especially those that deplete critical amino acids, have emerged as promising ones, some of which are already being used in clinical practice and many others are under development. This study reports the anti-cancer activity of two novel fused human arginase I (FHA) variants, FHA-3 and FHA-12, assessed using the NCI-60 human tumor cell line panel. Both variants have demonstrated a range of potencies in a single-dose assay (10 µM), but FHA-3 was found to be more potent with significant growth inhibition in most tested cell lines. To calculate 50% growth inhibition (GI50), FHA-3 was further evaluated in a five-dose assay, where notable anti-cancer activity was observed across the nine cancer types of the NCI-60 panel. Our results demonstrated the broad-spectrum anti-cancer activity of novel FHA variants, with FHA-3 being the most potent. Further studies elucidating its efficacy in animal models will help explore its therapeutic potential.
Collapse
Affiliation(s)
- Yenisetti Rajendra Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, Mohali, S.A.S. Nagar, 160062, Punjab, India
| | - J Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, Mohali, S.A.S. Nagar, 160062, Punjab, India
| | - Snehal Sainath Jawalekar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, Mohali, S.A.S. Nagar, 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, Mohali, S.A.S. Nagar, 160062, Punjab, India.
| |
Collapse
|
2
|
Jawalekar SS, Kawathe PS, Sharma N, Anakha J, Tikoo K, Pande AH. Development and characterization of fused human arginase I for cancer therapy. Invest New Drugs 2023; 41:652-663. [PMID: 37532976 DOI: 10.1007/s10637-023-01387-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
Recombinant human arginase I (rhArg I) have emerged as a potential candidate for the treatment of varied pathophysiological conditions ranging from arginine-auxotrophic cancer, inflammatory conditions and microbial infection. However, rhArg I have a low circulatory half-life, leading to poor pharmacokinetic and pharmacodynamic properties, which necessitating the rapid development of modifications to circumvent these limitations. To address this, polyethylene glycol (PEG)ylated-rhArg I variants are being developed by pharmaceutical companies. However, because of the limitations associated with the clinical use of PEGylated proteins, there is a dire need in the art to develop rhArg I variant(s) which is safe (devoid of limitations of PEGylated counterpart) and possess increased circulatory half-life. In this study, we described the generation and characterization of a fused human arginase I variant (FHA-3) having improved circulatory half-life. FHA-3 protein was engineered by fusing rhArg I with a half-life extension partner (domain of human serum albumin) via a peptide linker and was produced using P. pastoris expression system. This purified biopharmaceutical (FHA-3) exhibits (i) increased arginine-hydrolyzing activity in buffer, (ii) cofactor - independency, (iii) increased circulatory half-life (t1/2) and (iv) potent anti-cancer activity against human cancer cell lines under in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Snehal Sainath Jawalekar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Priyanka Sugriv Kawathe
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Nisha Sharma
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - J Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Kulbhushan Tikoo
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India.
| |
Collapse
|
3
|
Jiao YL, Shen PQ, Wang SF, Chen J, Zhou XH, Ma GZ. Arginase from Priestia megaterium and the Effects of CMCS Conjugation on Its Enzymological Properties. Curr Microbiol 2023; 80:292. [PMID: 37466752 DOI: 10.1007/s00284-023-03406-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023]
Abstract
Arginase has shown promising potential in treating cancers by arginine deprivation therapy; however, low enzymatic activity and stability of arginase are impeding its development. This study was aimed to improve the enzymological properties of a marine bacterial arginase by carboxymethyl chitosan (CMCS) conjugation. An arginase producing marine bacterium Priestia megaterium strain P6 was isolated and identified. The novel arginase PMA from the strain was heterologously expressed, purified, and then conjugated to CMCS by ionic gelation with calcium chloride as the crosslinking agent. Enzymological properties of both PMA and CMCS-PMA conjugate were determined. The optimum temperature for PMA and CMCS-PMA at pH 7 were 60 °C and 55 °C, respectively. The optimum pH for PMA and CMCS-PMA at 37 °C were pH 10 and 9, respectively. CMCS-PMA showed higher thermostability than PMA over 55-70 °C and higher pH stability over pH 4-11 with the highest pH stability at pH 7. At 37 °C and pH of 7, i.e., around the human blood temperature and pH, CMCS-PMA was higher than the free PMA in enzymatic activity and stability by 24% and 21%, respectively. CMCS conjugation not only changed the optimum temperature, optimum pH, and enzymatic activity of PMA, but also improved its pH stability and temperature stability, and thus made it more favorable for medical application.
Collapse
Affiliation(s)
- Yu Liang Jiao
- School of Marine Sciences and Fisheries, Jiangsu Ocean University, Cangwu Road, Lianyungang, Jiangsu, People's Republic of China.
| | - Pin Quan Shen
- School of Marine Sciences and Fisheries, Jiangsu Ocean University, Cangwu Road, Lianyungang, Jiangsu, People's Republic of China
| | - Shu Fang Wang
- School of Marine Sciences and Fisheries, Jiangsu Ocean University, Cangwu Road, Lianyungang, Jiangsu, People's Republic of China
| | - Jing Chen
- School of Marine Sciences and Fisheries, Jiangsu Ocean University, Cangwu Road, Lianyungang, Jiangsu, People's Republic of China
| | - Xiang Hong Zhou
- School of Marine Sciences and Fisheries, Jiangsu Ocean University, Cangwu Road, Lianyungang, Jiangsu, People's Republic of China
| | - Gui Zhen Ma
- School of Marine Sciences and Fisheries, Jiangsu Ocean University, Cangwu Road, Lianyungang, Jiangsu, People's Republic of China
| |
Collapse
|
4
|
Anakha J, Prasad YR, Sharma N, Pande AH. Human arginase I: a potential broad-spectrum anti-cancer agent. 3 Biotech 2023; 13:159. [PMID: 37152001 PMCID: PMC10156892 DOI: 10.1007/s13205-023-03590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/23/2023] [Indexed: 05/09/2023] Open
Abstract
With high rates of morbidity and mortality, cancer continues to pose a serious threat to public health on a global scale. Considering the discrepancies in metabolism between cancer and normal cells, metabolism-based anti-cancer biopharmaceuticals are gaining importance. Normal cells can synthesize arginine, but they can also take up extracellular arginine, making it a semi-essential amino acid. Arginine auxotrophy occurs when a cancer cell has abnormalities in the enzymes involved in arginine metabolism and relies primarily on extracellular arginine to support its biological functions. Taking advantage of arginine auxotrophy in cancer cells, arginine deprivation, which can be induced by introducing recombinant human arginase I (rhArg I), is being developed as a broad-spectrum anti-cancer therapy. This has led to the development of various rhArg I variants, which have shown remarkable anti-cancer activity. This article discusses the importance of arginine auxotrophy in cancer and different arginine-hydrolyzing enzymes that are in various stages of clinical development and reviews the need for a novel rhArg I that mitigates the limitations of the existing therapies. Further, we have also analyzed the necessity as well as the significance of using rhArg I to treat various arginine-auxotrophic cancers while considering the importance of their genetic profiles, particularly urea cycle enzymes.
Collapse
Affiliation(s)
- J. Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062 Punjab India
| | - Yenisetti Rajendra Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062 Punjab India
| | - Nisha Sharma
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062 Punjab India
| | - Abhay H. Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062 Punjab India
| |
Collapse
|
5
|
El Atat O, Naser R, Abdelkhalek M, Habib RA, El Sibai M. Molecular targeted therapy: A new avenue in glioblastoma treatment. Oncol Lett 2022; 25:46. [PMID: 36644133 PMCID: PMC9811647 DOI: 10.3892/ol.2022.13632] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma, also referred to as glioblastoma multiforme (GBM), is grade IV astrocytoma characterized by being fast-growing and the most aggressive brain tumor. In adults, it is the most prevalent type of malignant brain tumor. Despite the advancements in both diagnosis tools and therapeutic treatments, GBM is still associated with poor survival rate without any statistically significant improvement in the past three decades. Patient's genome signature is one of the key factors causing the development of this tumor, in addition to previous radiation exposure and other environmental factors. Researchers have identified genomic and subsequent molecular alterations affecting core pathways that trigger the malignant phenotype of this tumor. Targeting intrinsically altered molecules and pathways is seen as a novel avenue in GBM treatment. The present review shed light on signaling pathways and intrinsically altered molecules implicated in GBM development. It discussed the main challenges impeding successful GBM treatment, such as the blood brain barrier and tumor microenvironment (TME), the plasticity and heterogeneity of both GBM and TME and the glioblastoma stem cells. The present review also presented current advancements in GBM molecular targeted therapy in clinical trials. Profound and comprehensive understanding of molecular participants opens doors for innovative, more targeted and personalized GBM therapeutic modalities.
Collapse
Affiliation(s)
- Oula El Atat
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Rayan Naser
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Maya Abdelkhalek
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Ralph Abi Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Mirvat El Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon,Correspondence to: Professor Mirvat El Sibai, Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Koraytem Street, Beirut 1102 2801, Lebanon, E-mail:
| |
Collapse
|
6
|
Influence of the Nature of Amino Acids on the Formation of Supramolecular Complexes with Calixarenemethylene-bis-Phosphonic Acid. THEOR EXP CHEM+ 2022. [DOI: 10.1007/s11237-022-09742-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
7
|
Diwan D, Cheng L, Usmani Z, Sharma M, Holden N, Willoughby N, Sangwan N, Baadhe RR, Liu C, Gupta VK. Microbial cancer therapeutics: A promising approach. Semin Cancer Biol 2022; 86:931-950. [PMID: 33979677 DOI: 10.1016/j.semcancer.2021.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 01/27/2023]
Abstract
The success of conventional cancer therapeutics is hindered by associated dreadful side-effects of antibiotic resistance and the dearth of antitumor drugs' selectivity and specificity. Hence, the conceptual evolution of anti-cancerous therapeutic agents that selectively target cancer cells without impacting the healthy cells or tissues, has led to a new wave of scientific interest in microbial-derived bioactive molecules. Such strategic solutions may pave the way to surmount the shortcomings of conventional therapies and raise the potential and hope for the cure of wide range of cancer in a selective manner. This review aims to provide a comprehensive summary of anti-carcinogenic properties and underlying mechanisms of bioactive molecules of microbial origin, and discuss the current challenges and effective therapeutic application of combinatorial strategies to attain minimal systemic side-effects.
Collapse
Affiliation(s)
- Deepti Diwan
- Washington University, School of Medicine, Saint Louis, MO, USA
| | - Lei Cheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 230032, China
| | - Zeba Usmani
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Minaxi Sharma
- Department of Food Technology, Akal College of Agriculture, Eternal University, Baru Sahib, Himachal Pradesh, 173101, India
| | - Nicola Holden
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK
| | - Nicholas Willoughby
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Neelam Sangwan
- Department of Biochemistry, Central University of Haryana, Mahendergarh, Haryana, 123031, India
| | - Rama Raju Baadhe
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Vijai Kumar Gupta
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK; Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK.
| |
Collapse
|
8
|
Hou X, Chen S, Zhang P, Guo D, Wang B. Targeted Arginine Metabolism Therapy: A Dilemma in Glioma Treatment. Front Oncol 2022; 12:938847. [PMID: 35898872 PMCID: PMC9313538 DOI: 10.3389/fonc.2022.938847] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022] Open
Abstract
Efforts in the treatment of glioma which is the most common primary malignant tumor of the central nervous system, have not shown satisfactory results despite a comprehensive treatment model that combines various treatment methods, including immunotherapy. Cellular metabolism is a determinant of the viability and function of cancer cells as well as immune cells, and the interplay of immune regulation and metabolic reprogramming in tumors has become an active area of research in recent years. From the perspective of metabolism and immunity in the glioma microenvironment, we elaborated on arginine metabolic reprogramming in glioma cells, which leads to a decrease in arginine levels in the tumor microenvironment. Reduced arginine availability significantly inhibits the proliferation, activation, and function of T cells, thereby promoting the establishment of an immunosuppressive microenvironment. Therefore, replenishment of arginine levels to enhance the anti-tumor activity of T cells is a promising strategy for the treatment of glioma. However, due to the lack of expression of argininosuccinate synthase, gliomas are unable to synthesize arginine; thus, they are highly dependent on the availability of arginine in the extracellular environment. This metabolic weakness of glioma has been utilized by researchers to develop arginine deprivation therapy, which ‘starves’ tumor cells by consuming large amounts of arginine in circulation. Although it has shown good results, this treatment modality that targets arginine metabolism in glioma is controversial. Exploiting a suitable strategy that can not only enhance the antitumor immune response, but also “starve” tumor cells by regulating arginine metabolism to cure glioma will be promising.
Collapse
|
9
|
Chung SF, Tam SY, Kim CF, Chong HC, Lee LMY, Leung YC. Mono-PEGylated thermostable Bacillus caldovelox arginase mutant (BCA-M-PEG20) induces apoptosis, autophagy, cell cycle arrest and growth inhibition in gastric cancer cells. Invest New Drugs 2022; 40:895-904. [PMID: 35857203 PMCID: PMC9395487 DOI: 10.1007/s10637-022-01265-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/31/2022] [Indexed: 12/15/2022]
Abstract
Gastric cancer is one of the most common malignant solid tumors in the world, especially in Asia with high mortality due to a lack of effective treatment. The potential usage of the newly constructed arginine-depleting enzyme-mono-PEGylated Bacillus caldovelox arginase mutant (BCA-M-PEG20), an effective drug against multiple cancer cell lines such as cervical and lung cancers, for the treatment of gastric cancer was demonstrated. Our results indicated that BCA-M-PEG20 significantly inhibited argininosuccinate synthetase (ASS)-positive gastric cancer cells, MKN-45 and BGC-823, while another arginine-depleting enzyme, arginine deiminase (ADI, currently under Phase III clinical trial), failed to suppress the growth of gastric cancer cells. In vitro studies demonstrated that BCA-M-PEG20 inhibited MKN-45 cells by inducing autophagy and cell cycle arrest at the S phase under 0.58 U/mL (IC<sub>50</sub> values). Significant caspase-dependent apoptosis was induced in MKN-45 after the treatment with 2.32 U/mL of BCA-M-PEG20. In vivo studies showed that administrations of BCA-M-PEG20 at 250 U/mouse twice per week significantly suppressed about 50% of tumor growth in the MKN-45 gastric cancer xenograft model. Taken together, BCA-M-PEG20 demonstrated a superior potential to be an anti-gastric cancer drug.
Collapse
Affiliation(s)
- Sai-Fung Chung
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Centre 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
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Centre 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
| | - Chi-Fai Kim
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Centre 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
| | - Hiu-Chi Chong
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Centre 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
| | - Leo Man-Yuen Lee
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Centre 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
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Lo Ka Chung Research Centre 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
| |
Collapse
|
10
|
Zhu L, Zhou H, Xu F, Yang H, Li P, Sheng Y, Liu P, Kong W, Liu X, Yang L, Liu L, Liu X. Hepatic Ischemia-Reperfusion Impairs Blood-Brain Barrier Partly Due to Release of Arginase From Injured Liver. Front Pharmacol 2021; 12:724471. [PMID: 34721021 PMCID: PMC8548691 DOI: 10.3389/fphar.2021.724471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/13/2021] [Indexed: 11/20/2022] Open
Abstract
Aim: Hepatic ischemia-reperfusion (HIR) induces remote organs injury, including the brain. The homeostasis of the brain is maintained by the blood-brain barrier (BBB); thus, we aimed to investigate whether HIR impaired BBB and attempted to elucidate its underlying mechanism. Methods: Cell viability of human cerebral microvascular endothelial cells (hCMEC/D3) was measured following 24 h incubation with a serum of HIR rat undergoing 1 h ischemia and 4 h reperfusion, liver homogenate, or lysate of primary hepatocytes of the rat. The liver homogenate was precipitated using (NH4)2SO4 followed by separation on three columns and electrophoresis to identify the toxic molecule. Cell activity, apoptosis, proliferation, cell cycle, and expressions of proteins related to cell cycle were measured in hCMEC/D3 cells incubated with identified toxic molecules. HIR rats undergoing 1 h ischemia and 24 h reperfusion were developed to determine the release of an identified toxic molecule. BBB function was indexed as permeability to fluorescein and brain water. Endothelial cell proliferation and expressions of proteins related to the cell cycle in cerebral microvessels were measured by immunofluorescence and western blot. Results: Toxic molecule to BBB in the liver was identified to be arginase. Arginase inhibitor nor-NOHA efficiently attenuated hCMEC/D3 damage caused by liver homogenate and serum of HIR rats. Both arginase and serum of HIR rats significantly lowered arginine (Arg) in the culture medium. Arg addition efficiently attenuated the impairment of hCMEC/D3 caused by arginase or Arg deficiency, demonstrating that arginase impaired hCMEC/D3 via depriving Arg. Both arginase and Arg deficiency damaged hCMEC/D3 cells by inhibiting cell proliferation, retarding the cell cycle to G1 phase, and downregulating expressions of cyclin A, cyclin D, CDK2, and CDK4. HIR notably increased plasma arginase activity and lowered Arg level, increased the BBB permeability accompanied with enhanced brain water, and decreased the proliferative cells (marked by Ki67) in cerebral microvessels (marked by CD31) and protein expressions of cyclin A, cyclin D, CDK2 and CDK4 in isolated brain microvessels. Oral supplement of Arg remarkably attenuated these HIR-induced alterations. Conclusion: HIR leads to substantial release of arginase from the injured liver and then deprives systemic Arg. The Arg deficiency further impairs BBB via inhibiting the proliferation of brain microvascular endothelial cells by cell cycle arrest.
Collapse
Affiliation(s)
- Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Han Zhou
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng Xu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanyu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ping Li
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yun Sheng
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Peihua Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Weimin Kong
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaonan Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Wilder CS, Chen Z, DiGiovanni J. Pharmacologic approaches to amino acid depletion for cancer therapy. Mol Carcinog 2021; 61:127-152. [PMID: 34534385 DOI: 10.1002/mc.23349] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 11/09/2022]
Abstract
Cancer cells undergo metabolic reprogramming to support increased demands in bioenergetics and biosynthesis and to maintain reactive oxygen species at optimum levels. As metabolic alterations are broadly observed across many cancer types, metabolic reprogramming is considered a hallmark of cancer. A metabolic alteration commonly seen in cancer cells is an increased demand for certain amino acids. Amino acids are involved in a wide range of cellular functions, including proliferation, redox balance, bioenergetic and biosynthesis support, and homeostatic functions. Thus, targeting amino acid dependency in cancer is an attractive strategy for a number of cancers. In particular, pharmacologically mediated amino acid depletion has been evaluated as a cancer treatment option for several cancers. Amino acids that have been investigated for the feasibility of drug-induced depletion in preclinical and clinical studies for cancer treatment include arginine, asparagine, cysteine, glutamine, lysine, and methionine. In this review, we will summarize the status of current research on pharmacologically mediated amino acid depletion as a strategy for cancer treatment and potential chemotherapeutic combinations that synergize with amino acid depletion to further inhibit tumor growth and progression.
Collapse
Affiliation(s)
- Carly S Wilder
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Zhao Chen
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA.,Center for Molecular Carcinogenesis and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
12
|
El-Mais N, Fakhoury I, Al Haddad M, Nohra S, Abi-Habib R, El-Sibai M. Human Recombinant Arginase I [HuArgI(Co)-PEG5000]-Induced Arginine Depletion Inhibits Pancreatic Cancer Cell Migration and Invasion Through Autophagy. Pancreas 2021; 50:1187-1194. [PMID: 34714283 DOI: 10.1097/mpa.0000000000001891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Pancreatic cancer is one of the most aggressive solid cancers and the fourth leading cause of cancer death in men and women. We previously showed that arginine depletion, using arginase I [HuArgI(Co)-PEG5000], selectively triggers cell death by autophagy in PANC-1 pancreatic cancer cells. The mechanism of action of [HuArgI(Co)-PEG5000], however, has remained poorly understood. In this study, we investigated the effects of arginine depletion on PANC-1 cell migration, adhesion, and invasion and determined the main molecular targets, which mediate PANC-1 cell response to treatment with HuArgI(Co)-PEG5000. METHODS This was done through examining 2-dimensional (2D) cell motility assays (wound healing and time lapse), cell adhesion, and cell invasion assays, as well as immunostaining for focal adhesions and invadopodia in cells without or with the treatment with arginase. RESULTS We demonstrate that arginine depletion decreases PANC-1 2D cell migration, adhesion, and 3D invasion. Moreover, our data suggest that these effects are mediated by autophagy and subsequent decrease in the activation of members of Ras homolog gene family (Rho) GTPase family. CONCLUSIONS Altogether, these findings uncover the mechanism of action of [HuArgI(Co)-PEG5000] and highlight the promising and selective anticancer potential for arginine depletion in the treatment of pancreatic cancer cells.
Collapse
Affiliation(s)
- Nour El-Mais
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Isabelle Fakhoury
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Maria Al Haddad
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Sarah Nohra
- Department of Biosciences, School of Science and Technology, Università degli Studi di Milano, Milan, Italy
| | - Ralph Abi-Habib
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Mirvat El-Sibai
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
13
|
Kumari N, Bansal S. Arginine depriving enzymes: applications as emerging therapeutics in cancer treatment. Cancer Chemother Pharmacol 2021; 88:565-594. [PMID: 34309734 DOI: 10.1007/s00280-021-04335-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the second leading cause of death globally. Chemotherapy and radiation therapy and other medications are employed to treat various types of cancer. However, each treatment has its own set of side effects, owing to its low specificity. As a result, there is an urgent need for newer therapeutics that do not disrupt healthy cells' normal functioning. Depriving nutrient or non/semi-essential amino acids to which cancerous cells are auxotrophic remains one such promising anticancer strategy. L-Arginine (Arg) is a semi-essential vital amino acid involved in versatile metabolic processes, signaling pathways, and cancer cell proliferation. Hence, the administration of Arg depriving enzymes (ADE) such as arginase, arginine decarboxylase (ADC), and arginine deiminase (ADI) could be effective in cancer therapy. The Arg auxotrophic cancerous cells like hepatocellular carcinoma, human colon cancer, leukemia, and breast cancer cells are sensitive to ADE treatment due to low expression of crucial enzymes argininosuccinate synthetase (ASS), argininosuccinate lyase (ASL), and ornithine transcarbamylase (OCT). These therapeutic enzyme treatments induce cell death through inducing autophagy, apoptosis, generation of oxidative species, i.e., oxidative stress, and arresting the progression and expansion of cancerous cells at certain cell cycle checkpoints. The enzymes are undergoing clinical trials and could be successfully exploited as potential anticancer agents in the future.
Collapse
Affiliation(s)
- Neha Kumari
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology Waknaghat, Solan, 173234, Himachal Pradesh, India
| | - Saurabh Bansal
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology Waknaghat, Solan, 173234, Himachal Pradesh, India.
| |
Collapse
|
14
|
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
|
15
|
Swayden M, Bekdash A, Fakhoury I, El-Atat O, Borjac-Natour J, El-Sibai M, Abi-Habib RJ. Activation of autophagy following [HuArgI (Co)-PEG5000]-induced arginine deprivation mediates cell death in colon cancer cells. Hum Cell 2020; 34:152-164. [PMID: 32979152 DOI: 10.1007/s13577-020-00437-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023]
Abstract
Deregulating cellular energetics by reprogramming metabolic pathways, including arginine metabolism, is critical for cancer cell onset and survival. Drugs that target the specific metabolic requirements of cancer cells have emerged as promising targeted cancer therapeutics. In this study, we investigate the therapeutic potential of targeting colon cancer cells using arginine deprivation induced by a pegylated cobalt-substituted recombinant human Arginase I [HuArgI (Co)-PEG5000]. Four colon cancer cell lines were tested for their sensitivity to [HuArgI (Co)-PEG5000] as well as for their mechanism of cell death following arginine deprivation. All four cell lines were sensitive to arginine deprivation induced by [HuArgI (Co)-PEG5000]. All cells expressed ASS1 and were rescued from arginine deprivation-induced cytotoxicity by the addition of excess L-citrulline, indicating they are partially auxotrophic for arginine. Mechanistically, cells treated with [HuArgI (Co)-PEG5000] were negative for AnnexinV and lacked caspase activation. Further investigation revealed that arginine deprivation leads to a marked and prolonged activation of autophagy in both Caco-2 and T84 cell lines. Finally, we show that [HuArgI (Co)-PEG5000] causes cell death by sustained activation of autophagy as evidenced by the decrease in cell cytotoxicity upon treatment with chloroquine, an autophagy inhibitor. Altogether, these data demonstrate that colon cancer cells are partially auxotrophic for arginine and sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation. They also show that the activation of autophagy does not play protective roles but rather, induces cytotoxicity and leads to cell death.
Collapse
Affiliation(s)
- Mirna Swayden
- Department of Biological and Environmental Sciences, School of Arts and Sciences, Beirut Arab University, Beirut, Lebanon
| | - Amira Bekdash
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Isabelle Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Oula El-Atat
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Jamila Borjac-Natour
- Department of Biological and Environmental Sciences, School of Arts and Sciences, Beirut Arab University, Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Ralph J Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon.
| |
Collapse
|
16
|
Zhou W, Zeng X, Wu X. Effect of Oleanolic Acid on Apoptosis and Autophagy of SMMC-7721 Hepatoma Cells. Med Sci Monit 2020; 26:e921606. [PMID: 32424110 PMCID: PMC7251962 DOI: 10.12659/msm.921606] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Liver cancer is a common cancer with high morbidity and mortality. Due to the large toxic side effects of chemotherapeutic drugs and the overexpression of multidrug resistance genes in liver cancer, no effective chemotherapeutic drug has yet been found. Therefore, the search for a highly effective, low-toxic, and safe natural anticancer therapy is a hot issue. MATERIAL AND METHODS SMMC-7721 cells (a hepatocellular carcinoma cell line) were treated with different concentrations of oleanolic acid (OA) plus autophagy inhibitor 3-methyladenine (3-MA) (3-MA+OA) or chloroquine (CQ) plus OA (CQ+OA). We used MTT and Hoechst 33258 staining methods to determine the proliferation and apoptotic effect of OA on cells. Flow cytometry was used to detect apoptosis. Mitochondrial function was assessed by measuring mitochondrial membrane potential and adenosine triphosphate (ATP) concentration. To evaluate the ability of OA on apoptosis and autophagy mechanisms on SMMC 7721 cells, the related protein expression for apoptosis, autophagy, and the autophagic pathway were detected and analyzed by western blot. RESULTS OA can inhibit and induce apoptosis of SMMC-7721 in a dose-dependent manner. Compared with the control group, OA significantly reduced the intracellular mitochondrial membrane potential, and the intracellular ATP concentration was also significantly reduced. Moreover, OA reduced the expression of p-Akt and p-mTOR. The expression of p62 was decreased, and LC3-II and Beclin-1 protein expression levels increased. After inhibiting autophagy with 3-MA or CQ, compared with OA alone, cell mitochondrial membrane potential and ATP concentration were significantly reduced, cell p62 expression was reduced, and LC3-II expression was increased, apoptosis-related protein Bax protein was increased, and Bcl-2 protein was decreased, which suggested that 3-MA or CQ treatment increased OA-induced apoptosis of SMMC-7721 cells. This suggested that OA activated autophagy of SMMC-7721 cells in a protective autophagic manner. CONCLUSIONS The study findings suggest that OA combined with autophagy inhibitor 3-MA can better exert its anticancer effect.
Collapse
Affiliation(s)
- Weipeng Zhou
- The First Clinical Medical College of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Xianjun Zeng
- The First Afliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Xiaoping Wu
- The First Afliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
17
|
Al-Koussa H, El Mais N, Maalouf H, Abi-Habib R, El-Sibai M. Arginine deprivation: a potential therapeutic for cancer cell metastasis? A review. Cancer Cell Int 2020; 20:150. [PMID: 32390765 PMCID: PMC7201942 DOI: 10.1186/s12935-020-01232-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/23/2020] [Indexed: 01/04/2023] Open
Abstract
Arginine is a semi essential amino acid that is used in protein biosynthesis. It can be obtained from daily food intake or synthesized in the body through the urea cycle using l-citrulline as a substrate. Arginine has a versatile role in the body because it helps in cell division, wound healing, ammonia disposal, immune system, and hormone biosynthesis. It is noteworthy that l-arginine is the precursor for the biosynthesis of nitric oxide (NO) and polyamines. In the case of cancer cells, arginine de novo synthesis is not enough to compensate for their high nutritional needs, forcing them to rely on extracellular supply of arginine. In this review, we will go through the importance of arginine deprivation as a novel targeting therapy by discussing the different arginine deprivation agents and their mechanism of action. We will also focus on the factors that affect cell migration and on the influence of arginine on metastases through polyamine and NO.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Nour El Mais
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Hiba Maalouf
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Ralph Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| |
Collapse
|
18
|
Métayer LE, Brown RD, Carlebur S, Burke GAA, Brown GC. Mechanisms of cell death induced by arginase and asparaginase in precursor B-cell lymphoblasts. Apoptosis 2020; 24:145-156. [PMID: 30578463 PMCID: PMC6373273 DOI: 10.1007/s10495-018-1506-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Arginase has therapeutic potential as a cytotoxic agent in some cancers, but this is unclear for precursor B acute lymphoblastic leukaemia (pre-B ALL), the commonest form of childhood leukaemia. We compared arginase cytotoxicity with asparaginase, currently used in pre-B ALL treatment, and characterised the forms of cell death induced in a pre-B ALL cell line 697. Arginase and asparaginase both efficiently killed 697 cells and mature B lymphoma cell line Ramos, but neither enzyme killed normal lymphocytes. Arginase depleted cellular arginine, and arginase-treated media induced cell death, blocked by addition of arginine or arginine-precursor citrulline. Asparaginase depleted both asparagine and glutamine, and asparaginase-treated media induced cell death, blocked by asparagine, but not glutamine. Both enzymes induced caspase cleavage and activation, chromatin condensation and phosphatidylserine exposure, indicating apoptosis. Both arginase- and asparaginase-induced death were blocked by caspase inhibitors, but with different sensitivities. BCL-2 overexpression inhibited arginase- and asparaginase-induced cell death, but did not prevent arginase-induced cytostasis, indicating a different mechanism of growth arrest. An autophagy inhibitor, chloroquine, had no effect on the cell death induced by arginase, but doubled the cell death induced by asparaginase. In conclusion, arginase causes death of lymphoblasts by arginine-depletion induced apoptosis, via mechanism distinct from asparaginase. Therapeutic implications for childhood ALL include: arginase might be used as treatment (but antagonised by dietary arginine and citrulline), chloroquine may enhance efficacy of asparaginase treatment, and partial resistance to arginase and asparaginase may develop by BCL-2 expression. Arginase or asparaginase might potentially be used to treat Burkitt lymphoma.
Collapse
Affiliation(s)
- Lucy E Métayer
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Richard D Brown
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Saskia Carlebur
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - G A Amos Burke
- Department of Paediatrics, University of Cambridge, Cambridge, CB2 OQQ, UK
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| |
Collapse
|
19
|
A bioengineered arginine-depleting enzyme as a long-lasting therapeutic agent against cancer. Appl Microbiol Biotechnol 2020; 104:3921-3934. [DOI: 10.1007/s00253-020-10484-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/11/2020] [Accepted: 02/18/2020] [Indexed: 01/11/2023]
|
20
|
Khalil N, Abi-Habib RJ. [HuArgI (co)-PEG5000]-induced arginine deprivation leads to autophagy dependent cell death in pancreatic cancer cells. Invest New Drugs 2019; 38:1236-1246. [PMID: 31823161 DOI: 10.1007/s10637-019-00883-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/21/2019] [Indexed: 10/25/2022]
Abstract
In this study, we examined the sensitivity of pancreatic cancer cells to [HuArgI (Co)-PEG5000]-induced arginine deprivation as well as the mechanisms underlying deprivation-induced cell death. [HuArgI (Co)-PEG5000]-induced arginine deprivation was cytotoxic to all cell lines tested with IC50 values in the pM range at 72 h post-treatment. Three of the five cell lines were rescued by the addition of excess L-citrulline and expressed ASS1, indicating partial arginine auxotrophy. The remaining two cell lines, on the other hand, were not rescued by the addition of L-citrulline and did not express ASS1, indicating complete auxotrophy to arginine. In addition, all cell lines exhibited G0/G1 cell cycle arrest, in the surviving cell fraction, at 72 h following arginine deprivation. Analysis of the type of cell death revealed negative staining for annexin V and a lack of caspase activation in all five cancer cell lines, following treatment, indicating that arginine deprivation leads to caspase-independent, non-apoptotic cell death. Finally, we demonstrated that arginine deprivation leads to a marked activation of autophagy and that inhibition of this autophagy greatly decreases cytotoxicity, indicating that arginine deprivation induces autophagic cell death in pancreatic cancer cells. We have shown that pancreatic cancer cells are auxotrophic for arginine and sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation, hence demonstrating that arginine deprivation is a potentially potent and selective treatment for pancreatic cancer. We have also demonstrated that autophagy is activated following arginine-deprivation and that its prolonged activation leads to autophagic cell death.
Collapse
Affiliation(s)
- Nathalie Khalil
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Ralph J Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon.
| |
Collapse
|
21
|
Human Recombinant Arginase I [HuArgI (Co)-PEG5000]-Induced Arginine Depletion Inhibits Colorectal Cancer Cell Migration and Invasion. Int J Mol Sci 2019; 20:ijms20236018. [PMID: 31795337 PMCID: PMC6929075 DOI: 10.3390/ijms20236018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose: Colorectal cancer (CRC) is the third most common type of cancer worldwide, and it represents over half of all gastrointestinal cancer deaths. Knowing that cancer cells have a high proliferation rate, they require high amounts of amino acids, including arginine. In addition, several tumor types have been shown to downregulate ASS-1 expression, becoming auxotrophic for arginine. Therefore, Arginine deprivation is one of the promising therapeutic approaches to target cancer cells. This can be achieved through the use of a recombinant human arginase, HuArgI(Co)-PEG5000, an arginine degrading enzyme. Methods: In this present study, the cytotoxic effect of HuArgI(Co)-PEG5000 on CRC cell lines (HT-29, Caco-2, Sw837) is examined though cytotoxicity assays. Wound healing assays, invasion assays, and adhesion assays were also performed to detect the effect on metastasis. Results: Wound healing and invasion assays revealed a decrease in cell migration and invasion after treatment with arginase. Cells that were treated with arginase also showed a decrease in adhesion, which coincided with a decrease in RhoA activation, demonstrated though the use of a FRET biosensor to detect RhoA activation in a single cell assay, and a decrease in MMP-9 expression. Treating cells with both arginase and L-citrulline, which significantly restores intracellular arginine levels, reversed the effect of HuArgI(Co)-PEG5000 on cell viability, migration, and invasion. Conclusion: We can, therefore, conclude that colorectal cancer is partially auxotrophic to arginine and that arginine depletion is a potential selective inhibitory approach for motility and invasion in colon cancer cells.
Collapse
|
22
|
Nasreddine G, El-Sibai M, Abi-Habib RJ. Cytotoxicity of [HuArgI (co)-PEG5000]-induced arginine deprivation to ovarian Cancer cells is autophagy dependent. Invest New Drugs 2019; 38:10-19. [PMID: 30887252 DOI: 10.1007/s10637-019-00756-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/01/2019] [Indexed: 01/07/2023]
Abstract
In this study, we assess arginine auxotrophy in ovarian cancer cells and attempt to target them using arginine deprivation induced by a pegylated recombinant human Arginase I cobalt [HuArgI (Co)-PEG5000]. Ovarian cancer cells were sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation with IC50 values in the low pM range. Addition of excess L-citrulline rescued only one of three cell lines tested, indicating that the majority of cell lines are completely auxotrophic for arginine. The expression pattern of argininosuccinate synthetase (ASS1) confirmed the degree of auxotrophy of ovarian cancer cell lines with completely auxotrophic cells not expressing ASS1 and partially auxotrophic cells expressing the enzyme. Ovarian cancer cell lines were negative for annexinV staining while showing loss of membrane integrity and absence of caspase activation, indicating caspase-independent, non-apoptotic cell death. [HuArgI (Co)-PEG5000]-induced arginine deprivation led to extensive and prolonged activation of autophagy, which proved to be deleterious to cell survival since its inhibition led to a significant decrease in cytotoxicity. This indicates that the activation of autophagy following arginine-deprivation, rather than being protective, mediates cell cytotoxicity leading to death by autophagy.
Collapse
Affiliation(s)
- Ghenwa Nasreddine
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon
| | - Ralph J Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, 1102 2801, Lebanon.
| |
Collapse
|
23
|
Maggi M, Scotti C. Enzymes in Metabolic Anticancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:173-199. [PMID: 31482500 DOI: 10.1007/978-981-13-7709-9_9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancer treatment has greatly improved over the last 50 years, but it remains challenging in several cases. Useful therapeutic targets are normally unique peculiarities of cancer cells that distinguish them from normal cells and that can be tackled with appropriate drugs. It is now known that cell metabolism is rewired during tumorigenesis and metastasis as a consequence of oncogene activation and oncosuppressors inactivation, leading to a new cellular homeostasis typically directed towards anabolism. Because of these modifications, cells can become strongly or absolutely dependent on specific substrates, like sugars, lipids or amino acids. Cancer addictions are a relevant target for therapy, as removal of an essential substrate can lead to their selective cell-cycle arrest or even to cell death, leaving normal cells untouched. Enzymes can act as powerful agents in this respect, as demonstrated by asparaginase, which has been included in the treatment of Acute Lymphoblastic Leukemia for half a century. In this review, a short outline of cancer addictions will be provided, focusing on the main cancer amino acid dependencies described so far. Therapeutic enzymes which have been already experimented at the clinical level will be discussed, along with novel potential candidates that we propose as new promising molecules. The intrinsic limitations of their present molecular forms, along with molecular engineering solutions to explore, will also be presented.
Collapse
Affiliation(s)
- Maristella Maggi
- Department of Molecular Medicine, Unit of Immunology and General Pathology, University of Pavia, Pavia, Italy.
| | - Claudia Scotti
- Department of Molecular Medicine, Unit of Immunology and General Pathology, University of Pavia, Pavia, Italy
| |
Collapse
|
24
|
Conductometric biosensor for arginine determination in pharmaceutics. Bioelectrochemistry 2018; 124:40-46. [DOI: 10.1016/j.bioelechem.2018.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 06/25/2018] [Accepted: 07/03/2018] [Indexed: 11/17/2022]
|
25
|
Booth L, Roberts JL, Poklepovic A, Dent P. Prior exposure of pancreatic tumors to [sorafenib + vorinostat] enhances the efficacy of an anti-PD-1 antibody. Cancer Biol Ther 2018; 20:109-121. [PMID: 30142009 DOI: 10.1080/15384047.2018.1507258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Checkpoint immunotherapy antibodies have not shown efficacy in pancreatic adenocarcinoma. Pre-clinical studies and subsequently an on-going phase I trial have demonstrated the safety and efficacy of combinatorial radio-chemotherapy plus surgery in this malignancy, including the combination of sorafenib and vorinostat. The lethality of [sorafenib + vorinostat] was enhanced by gemcitabine. Exposure to [sorafenib + vorinostat] reduced the expression of β-catenin, ERBB1, BCL-XL and MCL-1, and the phosphorylation of AKT T308, AKT S473, GSK3 S9/21, mTORC1 and mTORC2. The drug combination increased the expression of Beclin1 and the phosphorylation of eIF2α S51. The drug combination rapidly reduced the levels of multiple HDAC proteins that was directly associated with the previously noted changes in tumor cell biology, as well as with alterations in the expression of biomarkers predictive for a response to checkpoint inhibitor antibodies. In vivo studies using the PAN02 model in its syngeneic mouse demonstrated that an anti-PD-1 antibody had no impact on tumor growth whereas a transient exposure to [sorafenib + vorinostat] significantly suppressed growth. The combination of [sorafenib + vorinostat] with an anti-PD-1 antibody caused a significant further reduction in tumor growth compared to the drug combination alone. Tumors transiently exposed three weeks earlier to [sorafenib + vorinostat] contained elevated levels of CD8+ cells, M1 macrophages and natural killer cells. Drug exposure plus an anti-PD-1 antibody further significantly enhanced the levels of these immune cells in the tumor. Our data argue for performing a new phase I trial in pancreatic cancer combining immunotherapy with [sorafenib + vorinostat]. Abbreviations: ERK: extracellular regulated kinase; PI3K: phosphatidyl inositol 3 kinase; ca: constitutively active; dn: dominant negative; ER: endoplasmic reticulum; AIF: apoptosis inducing factor; AMPK: AMP-dependent protein kinase; mTOR: mammalian target of rapamycin; JAK: Janus Kinase; STAT: Signal Transducers and Activators of Transcription; MAPK: mitogen activated protein kinase; PTEN: phosphatase and tensin homologue on chromosome ten; ROS: reactive oxygen species; CMV: empty vector plasmid or virus; si: small interfering; SCR: scrambled; IP: immunoprecipitation; VEH: vehicle; HDAC: histone deacetylase.
Collapse
Affiliation(s)
- Laurence Booth
- a Departments of Biochemistry and Molecular Biology , Virginia Commonwealth University , Richmond , VA , USA
| | - Jane Lisa Roberts
- a Departments of Biochemistry and Molecular Biology , Virginia Commonwealth University , Richmond , VA , USA
| | | | - Paul Dent
- a Departments of Biochemistry and Molecular Biology , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
26
|
Soldatkin OO, Marchenko SV, Soldatkina OV, Cherenok SO, Kalchenko OI, Prynova OS, Sylenko OM, Kalchenko VI, Dzyadevych SV. Conductometric sensor with calixarene-based chemosensitive element for the arginine detection. CHEMICAL PAPERS 2018. [DOI: 10.1007/s11696-018-0515-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
27
|
Affiliation(s)
- Wissam Zam
- Al-Andalus University for Medical Sciences, Syrian Arab Republic
| |
Collapse
|
28
|
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: 111] [Impact Index Per Article: 13.9] [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
|
29
|
Husain I, Bala K, Wani A, Makhdoomi U, Malik F, Sharma A. Arginase purified from endophytic Pseudomonas aeruginosa IH2: Induce apoptosis through both cell cycle arrest and MMP loss in human leukemic HL-60 cells. Chem Biol Interact 2017; 274:35-49. [PMID: 28690100 DOI: 10.1016/j.cbi.2017.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/20/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023]
Abstract
Arginase is a therapeutic enzyme for arginine-auxotrophic cancers but their low anticancer activity, less proteolytic tolerance and shorter serum half-life are the major shortcomings. In this study, arginase from Pseudomonas aeruginosa IH2 was purified to homogeneity and estimated as 75 kDa on native-PAGE and 37 kDa on SDS-PAGE. Arginase showed optimum activity at pH 8 and temperature 35 °C. Mn2+ and Mg2+ ions enhanced arginase activity while, Li+, Cu2+, and Al3+ ions reduced arginase activity. In-vitro serum half-life of arginase was 36 h and proteolytic half-life against trypsin and proteinase-K was 25 and 29 min, respectively. Anticancer activity of arginase was evaluated against colon, breast, leukemia, and prostate cancer cell lines and lowest IC50 (0.8 IU ml-1) was found against leukemia cell line HL-60. Microscopic studies and flow cytometric analysis of Annexin V/PI staining of HL-60 cells revealed that arginase induced apoptosis in dose-dependent manner. Cell cycle analysis suggested that arginase induced cell cycle arrest in G0/G1 phase. The increasing level of MMP loss, ROS generation and decreasing level of SOD, CAT, GPx and GSH suggested that arginase treatment triggered dysfunctioning of mitochondria. The cleavage of caspase-3, PARP-1, activations of caspase-8, 9 and high expression of proapoptotic protein Bax, low expression of anti-apoptotic protein Bcl-2 indicated that arginase treatment activates mitochondrial pathway of apoptosis. Purified arginase did not exert cytotoxic effects on human noncancer cells. Our study strongly supports that arginase could be used as potent anticancer agent but further studies are required which are underway in our lab.
Collapse
Affiliation(s)
- Islam Husain
- Bacteriology Laboratory, Department of P.G. Studies and Research in Biological Science, Rani Durgavati University, Madhya Pradesh, India
| | - Kiran Bala
- Bacteriology Laboratory, Department of P.G. Studies and Research in Biological Science, Rani Durgavati University, Madhya Pradesh, India
| | - Abubakar Wani
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu & Kashmir, India
| | - Ubaid Makhdoomi
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu & Kashmir, India
| | - Fayaz Malik
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu & Kashmir, India
| | - Anjana Sharma
- Bacteriology Laboratory, Department of P.G. Studies and Research in Biological Science, Rani Durgavati University, Madhya Pradesh, India.
| |
Collapse
|
30
|
Fernandes HS, Silva Teixeira CS, Fernandes PA, Ramos MJ, Cerqueira NMFSA. Amino acid deprivation using enzymes as a targeted therapy for cancer and viral infections. Expert Opin Ther Pat 2016; 27:283-297. [DOI: 10.1080/13543776.2017.1254194] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- H. S. Fernandes
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - C. S. Silva Teixeira
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - P. A. Fernandes
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - M. J. Ramos
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - N. M. F. S. A. Cerqueira
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| |
Collapse
|
31
|
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: 19.9] [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
|
32
|
Effects of the combined arginase and canavanine treatment on leukemic cells in vitro and in vivo. UKRAINIAN BIOCHEMICAL JOURNAL 2016; 88:45-55. [DOI: 10.15407/ubj88.02.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
33
|
Wang W, Iyer NG, Tay HT, Wu Y, Lim TKH, Zheng L, Song IC, Kwoh CK, Huynh H, Tan POB, Chow PKH. Microarray profiling shows distinct differences between primary tumors and commonly used preclinical models in hepatocellular carcinoma. BMC Cancer 2015; 15:828. [PMID: 26520397 PMCID: PMC4628260 DOI: 10.1186/s12885-015-1814-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite advances in therapeutics, outcomes for hepatocellular carcinoma (HCC) remain poor and there is an urgent need for efficacious systemic therapy. Unfortunately, drugs that are successful in preclinical studies often fail in the clinical setting, and we hypothesize that this is due to functional differences between primary tumors and commonly used preclinical models. In this study, we attempt to answer this question by comparing tumor morphology and gene expression profiles between primary tumors, xenografts and HCC cell lines. METHODS Hep G2 cell lines and tumor cells from patient tumor explants were subcutaneously (ectopically) injected into the flank and orthotopically into liver parenchyma of Mus Musculus SCID mice. The mice were euthanized after two weeks. RNA was extracted from the tumors, and gene expression profiling was performed using the Gene Chip Human Genome U133 Plus 2.0. Principal component analyses (PCA) and construction of dendrograms were conducted using Partek genomics suite. RESULTS PCA showed that the commonly used HepG2 cell line model and its xenograft counterparts were vastly different from all fresh primary tumors. Expression profiles of primary tumors were also significantly divergent from their counterpart patient-derived xenograft (PDX) models, regardless of the site of implantation. Xenografts from the same primary tumors were more likely to cluster together regardless of site of implantation, although heat maps showed distinct differences in gene expression profiles between orthotopic and ectopic models. CONCLUSIONS The data presented here challenges the utility of routinely used preclinical models. Models using HepG2 were vastly different from primary tumors and PDXs, suggesting that this is not clinically representative. Surprisingly, site of implantation (orthotopic versus ectopic) resulted in limited impact on gene expression profiles, and in both scenarios xenografts differed significantly from the original primary tumors, challenging the long-held notion that orthotopic PDX model is the gold standard preclinical model for HCC.
Collapse
Affiliation(s)
- Weining Wang
- Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore.
| | - N Gopalakrishna Iyer
- Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore. .,Department of Surgical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
| | - Hsien Ts'ung Tay
- Department of General Surgery, Singapore General Hospital, 11 Hospital Drive, Singapore, 169608, Singapore.
| | - Yonghui Wu
- Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore.
| | - Tony K H Lim
- Department of Histopathology, Singapore General Hospital, 11 Hospital Drive, Singapore, 169608, Singapore.
| | - Lin Zheng
- SingHealth Experimental Medicine Centre (SEMC), Blk 9, Level 3, Outram Road, Singapore, 169608, Singapore.
| | - In Chin Song
- SingHealth Experimental Medicine Centre (SEMC), Blk 9, Level 3, Outram Road, Singapore, 169608, Singapore.
| | - Chee Keong Kwoh
- Division of Information Systems, School of Computer Engineering, Nanyang Technological University, Nanyang Avenue, Singapore, 639798, Singapore.
| | - Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, 11 Hospital Drive, Singapore, 169610, Singapore.
| | - Patrick O B Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore, 169857, Singapore.
| | - Pierce K H Chow
- Department of Surgical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore. .,Program in Translational and Clinical Liver Research, National Cancer Centre Singapore, Singapore, 169610, Singapore. .,Office of Clinical Sciences, Duke-NUS Graduate Medical School, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
34
|
IL-10 and ARG-1 concentrations in bone marrow and peripheral blood of metastatic neuroblastoma patients do not associate with clinical outcome. J Immunol Res 2015; 2015:718975. [PMID: 25961062 PMCID: PMC4417583 DOI: 10.1155/2015/718975] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/25/2014] [Indexed: 12/22/2022] Open
Abstract
The expression of the immunosuppressive molecules IL-10 and arginase 1 (ARG-1), and of FOXP3 and CD163, as markers of regulatory T cells (Treg) and macrophages, respectively, was evaluated in bone marrow (BM) and peripheral blood (PB) samples collected at diagnosis from patients with metastatic neuroblastoma (NB). IL-10 and ARG-1 plasma concentrations were measured and the association of each parameter with patients' outcome was tested. The percentages of immunosuppressive Treg and type-1 regulatory (Tr1) cells were also determined. In both BM and PB samples, IL-10 mRNA expression was higher in metastatic NB patients than in controls. IL-10 plasma concentration was higher in patients with NB regardless of stage. Neither IL-10 expression nor IL-10 plasma concentration significantly associated with patient survival. In PB samples from metastatic NB patients, ARG-1 and CD163 expression was higher than in controls but their expression did not associate with survival. Moreover, ARG-1 plasma concentration was lower than in controls, and no association with patient outcome was found. Finally, in metastatic NB patients, the percentage of circulating Treg was higher than in controls, whereas that of Tr1 cells was lower. In conclusion, although IL-10 concentration and Treg percentage were increased, their contribution to the natural history of metastatic NB appears uncertain.
Collapse
|
35
|
Human recombinant arginase I (Co)-PEG5000 [HuArgI (Co)-PEG5000]-induced arginine depletion is selectively cytotoxic to human glioblastoma cells. J Neurooncol 2015; 122:75-85. [PMID: 25567351 DOI: 10.1007/s11060-014-1698-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
Abstract
In this study, we attempt to target Arginine auxotrophy in glioblastoma multiforme (GBM) cells using a pegylated recombinant human Arginase I cobalt [HuArgI (Co)-PEG5000]. We tested and characterized the activity of HuArgI (Co)-PEG5000 on a panel of 9 GBM cell lines and on human fetal glial cells (SVG-p12). HuArgI (Co)-PEG5000 was cytotoxic to all GBM cells tested. SVG-p12 cells were not sensitive demonstrating the selective cytotoxicity of HuArgI (Co)-PEG5000-induced arginine deprivation. Addition of L-citrulline led to the rescue of 6 GBM cell lines but only at concentrations of 11.4 mM, reflecting the extent of arginine auxotrophy in GBM. The ability of L-citrulline to rescue cells was dependent on the expression of argininosuccinate synthetase-1 (ASS1) with the cells that were not rescued by L-citrulline being negative for ASS1 expression. Knocking-down ASS1 reversed the ability of L-citrulline to rescue GBM cells, further illustrating the dependence of arginine auxotrophy on ASS1 expression. Inhibition of autophagy increased cell sensitivity to HuArgI (Co)-PEG5000 indicating that, following arginine deprivation, autophagy plays a protective role in GBM cells. Analysis of the type of cell death revealed a lack of AnnexinV staining and caspase activation in HuArgI (Co)-PEG5000-treated cells, indicating that arginine deprivation induces caspase-independent, non-apoptotic cell death in GBM. We have shown that GBM cells are auxotrophic for arginine and can be selectively targeted using HuArgI (Co)-PEG5000-induced arginine depletion, thus demonstrating that L-Arginine deprivation is a potent and selective potential treatment for GBM.
Collapse
|
36
|
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
|
37
|
Zeng X, Li Y, Fan J, Zhao H, Xian Z, Sun Y, Wang Z, Wang S, Zhang G, Ju D. Recombinant human arginase induced caspase-dependent apoptosis and autophagy in non-Hodgkin's lymphoma cells. Cell Death Dis 2013; 4:e840. [PMID: 24113174 PMCID: PMC3824669 DOI: 10.1038/cddis.2013.359] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/05/2013] [Accepted: 08/12/2013] [Indexed: 12/19/2022]
Abstract
Arginase, an arginine-degrading enzyme, has gained increased attention recently as a new experimental therapeutics for a variety of malignant solid cancers. In this study, we found that recombinant human arginase (rhArg) could induce remarkable growth inhibition, cell cycle arrest, and caspase-dependent apoptosis in Raji and Daudi non-Hodgkin's lymphoma (NHL) cells through arginine deprivation. Interestingly, rhArg-treatment resulted in the appearance of autophagosomes and upregulation of microtubule-associated protein light chain 3 II, indicating that rhArg induced autophagy in lymphoma cells. Further study suggested that mammalian target of rapamycin/S6k signaling pathway may be involved in rhArg-induced autophagy in NHL cells. Moreover, blocking autophagy using pharmacological inhibitors (3-methyladenine and chloroquine) or genetic approaches (small interfering RNA targeting autophagy-related gene 5 and Beclin-1) enhanced the cell killing effect of rhArg. These results demonstrated that rhArg has a potent anti-lymphoma activity, which could be improved by in combination with autophagic inhibitors, suggesting that rhArg, either alone or in combination with autophagic inhibitors, could be a potential novel therapeutics for the treatment of NHL.
Collapse
Affiliation(s)
- X Zeng
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai 201203, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Narayanan SP, Rojas M, Suwanpradid J, Toque HA, Caldwell RW, Caldwell RB. Arginase in retinopathy. Prog Retin Eye Res 2013; 36:260-80. [PMID: 23830845 PMCID: PMC3759622 DOI: 10.1016/j.preteyeres.2013.06.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/14/2013] [Accepted: 06/25/2013] [Indexed: 12/12/2022]
Abstract
Ischemic retinopathies, such as diabetic retinopathy (DR), retinopathy of prematurity and retinal vein occlusion are a major cause of blindness in developed nations worldwide. Each of these conditions is associated with early neurovascular dysfunction. However, conventional therapies target clinically significant macula edema or neovascularization, which occur much later. Intra-ocular injections of anti-VEGF show promise in reducing retinal edema, but the effects are usually transient and the need for repeated injections increases the risk of intraocular infection. Laser photocoagulation can control pathological neovascularization, but may impair vision and in some patients the retinopathy continues to progress. Moreover, neither treatment targets early stage disease or promotes repair. This review examines the potential role of the ureahydrolase enzyme arginase as a therapeutic target for the treatment of ischemic retinopathy. Arginase metabolizes l-arginine to form proline, polyamines and glutamate. Excessive arginase activity reduces the l-arginine supply for nitric oxide synthase (NOS), causing it to become uncoupled and produce superoxide and less NO. Superoxide and NO react and form the toxic oxidant peroxynitrite. The catabolic products of polyamine oxidation and glutamate can induce more oxidative stress and DNA damage, both of which can cause cellular injury. Studies indicate that neurovascular injury during retinopathy is associated with increased arginase expression/activity, decreased NO, polyamine oxidation, formation of superoxide and peroxynitrite and dysfunction and injury of both vascular and neural cells. Furthermore, data indicate that the cytosolic isoform arginase I (AI) is involved in hyperglycemia-induced dysfunction and injury of vascular endothelial cells whereas the mitochondrial isoform arginase II (AII) is involved in neurovascular dysfunction and death following hyperoxia exposure. Thus, we postulate that activation of the arginase pathway causes neurovascular injury by uncoupling NOS and inducing polyamine oxidation and glutamate formation, thereby reducing NO and increasing oxidative stress, all of which contribute to the retinopathic process.
Collapse
Affiliation(s)
- S. Priya Narayanan
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Modesto Rojas
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Jutamas Suwanpradid
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology & Toxicology, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - R. William Caldwell
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Pharmacology & Toxicology, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Ruth B. Caldwell
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- VA Medical Center, One Freedom Way, Augusta, GA, USA
| |
Collapse
|
40
|
Tanios R, Bekdash A, Kassab E, Stone E, Georgiou G, Frankel AE, Abi-Habib RJ. Human recombinant arginase I(Co)-PEG5000 [HuArgI(Co)-PEG5000]-induced arginine depletion is selectively cytotoxic to human acute myeloid leukemia cells. Leuk Res 2013; 37:1565-71. [PMID: 24018014 DOI: 10.1016/j.leukres.2013.08.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 01/14/2023]
Abstract
In this study, we target arginine auxotrophy of AML cell lines using human arginase I cobalt-PEG5000. HuArgI(Co)-PEG5000 was cytotoxic to all AML cell lines tested. Mononuclear cells and CD34(+) blasts were not sensitive demonstrating the selectivity of HuArgI(Co)-PEG5000-induced arginine deprivation. Addition of L-citrulline led to the rescue of five cell lines. The four cell lines that were not rescued by L-citrulline did not express argininosuccinate synthetase-1, indicating complete arginine auxotrophy. Inhibition of autophagy increased cell sensitivity to HuArgI(Co)-PEG5000 demonstrating the protective role of autophagy following arginine deprivation. We have shown that AML can be selectively targeted using HuArgI(Co)-PEG5000-induced arginine depletion.
Collapse
Affiliation(s)
- Rita Tanios
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | | | | | | | | | | | | |
Collapse
|
41
|
Srivastava A, Meena SK, Alam M, Nayeem SM, Deep S, Sau AK. Structural and Functional Insights into the Regulation of Helicobacter pylori Arginase Activity by an Evolutionary Nonconserved Motif. Biochemistry 2013; 52:508-19. [DOI: 10.1021/bi301421v] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Abhishek Srivastava
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110 067,
India
| | - Shiv Kumar Meena
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110 067,
India
| | - Mashkoor Alam
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110 067,
India
| | - Shahid M. Nayeem
- Department of Chemistry, Indian Institute of Technology, New Delhi 110 016,
India
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology, New Delhi 110 016,
India
| | - Apurba Kumar Sau
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110 067,
India
| |
Collapse
|
42
|
Yoon JK, Frankel AE, Feun LG, Ekmekcioglu S, Kim KB. Arginine deprivation therapy for malignant melanoma. Clin Pharmacol 2012; 5:11-9. [PMID: 23293541 PMCID: PMC3534294 DOI: 10.2147/cpaa.s37350] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Despite recent development of promising immunotherapeutic and targeted drugs, prognosis in patients with advanced melanoma remains poor, and a cure for this disease remains elusive in most patients. The success of melanoma therapy depends on a better understanding of the biology of melanoma and development of drugs that effectively target the relevant genes or proteins essential for tumor cell survival. Melanoma cells frequently lack argininosuccinate synthetase, an essential enzyme for arginine synthesis, and as a result they become dependent on the availability of exogenous arginine. Accordingly, a therapeutic approach involving depletion of available arginine has been shown to be effective in preclinical studies. Early clinical studies have demonstrated sufficient antitumor activity to give rise to cautious optimism. In this article, the rationale for arginine deprivation therapy is discussed. Additionally, various strategies for depleting arginine are discussed and the preclinical and clinical investigations of arginine deprivation therapy in melanoma are reviewed.
Collapse
Affiliation(s)
- Jung-Ki Yoon
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA ; Hwasung Public Health Center, Hwasung, South Korea
| | | | | | | | | |
Collapse
|
43
|
D’Antonio EL, Christianson DW. Binding of the unreactive substrate analog L-2-amino-3-guanidinopropionic acid (dinor-L-arginine) to human arginase I. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:889-93. [PMID: 22869115 PMCID: PMC3412766 DOI: 10.1107/s1744309112027820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 06/19/2012] [Indexed: 11/11/2022]
Abstract
Human arginase I (HAI) is a binuclear manganese metalloenzyme that catalyzes the hydrolysis of L-arginine to form L-ornithine and urea through a metal-activated hydroxide mechanism. Since HAI regulates L-Arg bioavailability for NO biosynthesis, it is a potential drug target for the treatment of cardiovascular diseases such as atherosclerosis. X-ray crystal structures are now reported of the complexes of Mn(2)(2+)-HAI and Co(2)(2+)-HAI with L-2-amino-3-guanidinopropionic acid (AGPA; also known as dinor-L-arginine), an amino acid bearing a guanidinium side chain two methylene groups shorter than that of L-arginine. Hydrogen bonds to the α-carboxylate and α-amino groups of AGPA dominate enzyme-inhibitor recognition; the guanidinium group does not interact directly with the metal ions.
Collapse
Affiliation(s)
- Edward L. D’Antonio
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - David W. Christianson
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| |
Collapse
|
44
|
Romero PA, Stone E, Lamb C, Chantranupong L, Krause A, Miklos AE, Hughes RA, Fechtel B, Ellington AD, Arnold FH, Georgiou G. SCHEMA-designed variants of human Arginase I and II reveal sequence elements important to stability and catalysis. ACS Synth Biol 2012; 1:221-8. [PMID: 22737599 DOI: 10.1021/sb300014t] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Arginases catalyze the divalent cation-dependent hydrolysis of L-arginine to urea and L-ornithine. There is significant interest in using arginase as a therapeutic antineogenic agent against L-arginine auxotrophic tumors and in enzyme replacement therapy for treating hyperargininemia. Both therapeutic applications require enzymes with sufficient stability under physiological conditions. To explore sequence elements that contribute to arginase stability we used SCHEMA-guided recombination to design a library of chimeric enzymes composed of sequence fragments from the two human isozymes Arginase I and II. We then developed a novel active learning algorithm that selects sequences from this library that are both highly informative and functional. Using high-throughput gene synthesis and our two-step active learning algorithm, we were able to rapidly create a small but highly informative set of seven enzymatically active chimeras that had an average variant distance of 40 mutations from the closest parent arginase. Within this set of sequences, linear regression was used to identify the sequence elements that contribute to the long-term stability of human arginase under physiological conditions. This approach revealed a striking correlation between the isoelectric point and the long-term stability of the enzyme to deactivation under physiological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Andreas Krause
- Department of Computer Science, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
45
|
Recombinant human arginase toxicity in mice is reduced by citrulline supplementation. Transl Oncol 2012; 5:26-31. [PMID: 22348173 DOI: 10.1593/tlo.11262] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 08/29/2011] [Accepted: 09/23/2011] [Indexed: 11/18/2022] Open
Abstract
Human recombinant arginase I cobalt coupled to polyethylene glycol 5000 (HuArg I [Co]-PEG5000) achieved potent in vitro depletion of arginine from tissue culture medium and cytotoxicity to many cancer cell lines. The recombinant enzyme also produced tumor growth inhibition of hepatocellular carcinoma and pancreatic carcinoma xenografts. Although these results were promising, the therapeutic index was narrow. Toxicities were seen in normal cells in tissue culture. In vivo normal tissue injury occurred at doses twice the effective dose. The current study was conducted to define, in greater detail, the maximum tolerated dose (MTD), pharmacodynamics, and dose-limiting toxicities (DLTs) of twice-weekly intraperitoneal HuArg I [Co]-PEG5000 in Balb/c mice. Animal weight and survival were monitored, serum arginine levels measured, and complete blood cell counts, chemistries, necropsies, and histologies were performed. In addition, methods to ameliorate the HuArg I [Co]-PEG5000 adverse effects were tested. Supplemental l-citrulline was given concurrently with the arginase drug. The HuArg I [Co]-PEG5000 MTD in mice was 5 mg/kg twice weekly, and DLTs included weight loss and marrow necrosis. No other organ damage or changes in blood cell counts or chemistries were observed. Arginase reduced serum arginine levels from 60 µM to 4 to 6 µM. Supplemental l-citrulline given per os or daily subcutaneously reduced and delayed toxicities, and l-citrulline given twice daily subcutaneously completely prevented animal toxicities. On the basis of these results, we hypothesize that HuArg I [Co]-PEG5000, particularly with supplemental l-citrulline, may be an attractive therapeutic agent for argininosuccinate synthetase-deficient tumors.
Collapse
|
46
|
Stone E, Chantranupong L, Gonzalez C, O'Neal J, Rani M, VanDenBerg C, Georgiou G. Strategies for optimizing the serum persistence of engineered human arginase I for cancer therapy. J Control Release 2011; 158:171-9. [PMID: 22001609 DOI: 10.1016/j.jconrel.2011.09.097] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/17/2011] [Accepted: 09/29/2011] [Indexed: 11/16/2022]
Abstract
Systemic L-arginine depletion following intravenous administration of l-arginine hydrolyzing enzymes has been shown to selectively impact tumors displaying urea cycle defects including a large fraction of hepatocellular carcinomas, metastatic melanomas and small cell lung carcinomas. However, the human arginases display poor serum stability (t(1/2)=4.8h) whereas a bacterial arginine deiminase evaluated in phase II clinical trials was reported to be immunogenic, eliciting strong neutralizing antibody responses. Recently, we showed that substitution of the Mn(2+) metal center in human Arginase I with Co(2+) (Co-hArgI) results in an enzyme that displays 10-fold higher catalytic efficiency for L-Arg hydrolysis, 12-15 fold reduction in the IC(50) towards a variety of malignant cell lines and, importantly a t(1/2)=22h in serum. To investigate the utility of Co-hArgI for L-Arg depletion therapy in cancer we systematically investigated three strategies for enhancing the persistence of the enzyme in circulation: (i) site specific conjugation of Co-hArgI engineered with an accessible N-terminal Cys residue to 20kDa PEG-maleimide (Co-hArgI-C(PEG-20K)); (ii) engineering of the homotrimeric Co-hArgI into a linked, monomeric 110kDa polypeptide (Co-hArgI x3) and (iii) lysyl conjugation of 5kDa PEG-N-hydroxysuccinimide (NHS) ester (Co-hArgI-K(PEG-5K)). Surprisingly, even though all three formulations resulted in proteins with a predicted hydrodynamic radius larger than the cut-off for renal filtration, only Co-hArgI amine conjugated to 5kDa PEG remained in circulation for sufficiently long durations. Using Co-hArgI-K(PEG-5K) labeled with an end-terminal fluorescein for easy detection, we demonstrated that following intraperitoneal administration at 6mg/kg weight, a well tolerated dose, the circulation t(1/2) of the protein in Balb/c mice is 63±10h. Very low levels of serum L-Arg (<5μM) could be sustained for over 75h after injection, representing a 9-fold increase in pharmacodynamic efficacy relative to similarly prepared Mn(2+)-containing hArgI conjugated to 5kDa PEG-NHS ester (Mn-hArgI-K(PEG-5K)). The favorable pharmacokinetic and pharmacodynamic properties of Co-hArgI-K(PEG-5K) reported here, coupled with its human origin which should reduce the likelihood of adverse immune responses, make it a promising candidate for cancer therapy.
Collapse
Affiliation(s)
- Everett Stone
- Department of Chemical Engineering, Center for Molecular, College of Pharmacy, University of Texas, Austin, TX 78712, USA
| | | | | | | | | | | | | |
Collapse
|