Open Access Open Access  Restricted Access Subscription or Fee Access

Human Arginase I (Arg I)—A Potential Broad-spectrum Anti-cancer Agent: Perspectives and the Road Ahead

Sayantap Datta, Priyanka Kawathe, Snehal Sainath Jawalekar, Abhay H Pande

Abstract


Cancer is the second leading cause of morbidity and mortality globally. A variety of effective regimens are available for the treatment of different cancers, however, there is still a need for the development of safe and effective broad-spectrum anti-cancer agent(s). Towards this, amino acid-depleting enzymes have proven to be effective in the treatment of certain auxotrophic cancers. In this review, we have briefly discussed arginine metabolism and arginine depletion as an effective strategy in the treatment of arginine-auxotrophic cancers. We have discussed the latest events in the field of development of recombinant human Arg I (rh-Arg I) for different cancers, current status of development of various arginine-depleting enzymes, their limitations, and potential approaches to overcome these limitations.

 

Keywords: Auxotrophy, arginine metabolism, arginine deiminase, arginase, fusion protein technology

 

Cite this Article

Sayantap Datta, Priyanka Kawathe, Snehal Sainath Jawalekar, Abhay H Pande. Human Arginase I (Arg I)—A Potential Broad-spectrum Anti-cancer Agent: Perspectives and the Road Ahead. Research & Reviews: Journal of Oncology and Hematology. 2020; 9(3): 23–35p. DOI: https://doi.org/10.37591/rrjooh.v9i3.2312


Full Text:

PDF

References


Research, I.C.o.M., Indian Council of Medical Research Media Report (2 February to 8 February 2019) (ICMR IN NEWS). 2019.

Gupta, G.P. and J. Massagué, Cancer metastasis: building a framework. Cell, 2006. 127(4): p. 679–95.

Moore, S.C., et al., Association of Leisure-Time Physical Activity With Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Intern Med, 2016. 176(6): p. 816–25.

Organization, W.H., Cancer. 2020.

Research, I.C.o.M., Cancer Statistics. 2020.

Ng, E., J. Dong, and D. Ratner, Basal Cell Carcinoma, in Evidence-Based Procedural Dermatology. 2019, Springer. p. 723–748.

Wang, T.H., et al., Combined Yttrium-90 microsphere selective internal radiation therapy and external beam radiotherapy in patients with hepatocellular carcinoma: From clinical aspects to dosimetry. PLoS One, 2018. 13(1): p. e0190098.

Society, A.C., How Chemotherapy Drugs Work. 2019.

Zhou, L., et al., Targeted biopharmaceuticals for cancer treatment. Cancer Lett, 2014. 352(2): p. 145–51.

Andersen, J.T., et al., Extending serum half-life of albumin by engineering neonatal Fc receptor (FcRn) binding. J Biol Chem, 2014. 289(19): p. 13492–502.

Patil, M.D., et al., Arginine dependence of tumor cells: targeting a chink in cancer's armor. Oncogene, 2016. 35(38): p. 4957–72.

van Niekerk, G., T. Nell, and A.M. Engelbrecht, Domesticating Cancer: An Evolutionary Strategy in the War on Cancer. Front Oncol, 2017. 7: p. 304.

Guedes, R., et al. Amino acids biosynthesis and nitrogen assimilation pathways: a great genomic deletion during eukaryotes evolution. in BMC genomics. 2011. Springer.

Cantor, J.R., et al., Engineering reduced-immunogenicity enzymes for amino acid depletion therapy in cancer. Methods Enzymol, 2012. 502: p. 291–319.

Jeon, H., et al., Methionine deprivation suppresses triple-negative breast cancer metastasis in vitro and in vivo. Oncotarget, 2016. 7(41): p. 67223–67234.

Hoffman, R.M., Development of recombinant methioninase to target the general cancer-specific metabolic defect of methionine dependence: a 40-year odyssey. Expert Opin Biol Ther, 2015. 15(1): p. 21–31.

Tan, Y., et al., Serum methionine depletion without side effects by methioninase in metastatic breast cancer patients. Anticancer Res, 1996. 16(6c): p. 3937–42.

Kahraman, H., Pseudomonas aeruginosa expressing Vitreoscilla hemoglobin shows increased production of L-lysine α-oxidase: an enzyme used in cancer therapy. Fırat University Turkish Journal of Science and Technology, 2018. 13(2): p. 47–52.

Treshalina, H.M., et al., Anticancer enzyme L-lysine α-oxidase. Applied biochemistry and biotechnology, 2000. 88(1–3): p. 267–273.

Biotherapeutics, A., Providing Solutions for Diseases With Unmet Medical Need. 2020.

Morris, S.M., Jr., Arginine: beyond protein. Am J Clin Nutr, 2006. 83(2): p. 508s-512s.

Stasyuk, N., et al., Fluorometric enzymatic assay of l-arginine. Spectrochim Acta A Mol Biomol Spectrosc, 2017. 170: p. 184–90.

Lam, S.K., et al., Inhibition of ornithine decarboxylase 1 facilitates pegylated arginase treatment in lung adenocarcinoma xenograft models. Oncol Rep, 2018. 40(4): p. 1994–2004.

Delage, B., et al., Promoter methylation of argininosuccinate synthetase-1 sensitises lymphomas to arginine deiminase treatment, autophagy and caspase-dependent apoptosis. Cell death & disease, 2012. 3(7): p. e342-e342.

Tsai, W.-B., et al., Resistance to arginine deiminase treatment in melanoma cells is associated with induced argininosuccinate synthetase expression involving c-Myc/HIF-1α/Sp4. Molecular cancer therapeutics, 2009. 8(12): p. 3223–3233.

Delage, B., et al., Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. International journal of cancer, 2010. 126(12): p. 2762–2772.

Alexandrou, C., et al., Sensitivity of Colorectal Cancer to Arginine Deprivation Therapy is Shaped by Differential Expression of Urea Cycle Enzymes. Sci Rep, 2018. 8(1): p. 12096.

O'Dwyer, D., et al., The proteomics of colorectal cancer: identification of a protein signature associated with prognosis. PLoS One, 2011. 6(11): p. e27718.

Wang, M., et al., Engineering an arginine catabolizing bioconjugate: biochemical and pharmacological characterization of PEGylated derivatives of arginine deiminase from Mycoplasma arthritidis. Bioconjugate chemistry, 2006. 17(6): p. 1447–1459.

Szlosarek, P.W., et al., In vivo loss of expression of argininosuccinate synthetase in malignant pleural mesothelioma is a biomarker for susceptibility to arginine depletion. Clin Cancer Res, 2006. 12(23): p. 7126–31.

Lind, D.S., Arginine and cancer. J Nutr, 2004. 134(10 Suppl): p. 2837S-2841S; discussion 2853S.

Kim, R.H., et al., Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res, 2009. 69(2): p. 700–8.

Dhankhar, R., et al., Arginine-lowering enzymes against cancer: a technocommercial analysis through patent landscape. Expert Opinion on Therapeutic Patents, 2018. 28(8): p. 603–614.

Bobak, Y., et al., Arginine deprivation induces endoplasmic reticulum stress in human solid cancer cells. The international journal of biochemistry & cell biology, 2016. 70: p. 29–38.

Polaris, Leading the way to new and better cancer treatments. 2020.

Feun, L. and N. Savaraj, Pegylated arginine deiminase: a novel anticancer enzyme agent. Expert Opin Investig Drugs, 2006. 15(7): p. 815–22.

Yoon, J.K., et al., Arginine deprivation therapy for malignant melanoma. Clin Pharmacol, 2013. 5: p. 11–9.

Fung, M.K.L. and G.C. Chan, Drug-induced amino acid deprivation as strategy for cancer therapy. J Hematol Oncol, 2017. 10(1): p. 144.

Wong, B.L., et al., Albumin-binding arginine deminase and the use thereof. 2016, Google Patents.

Caldwell, R.B., et al., Arginase: an old enzyme with new tricks. Trends Pharmacol Sci, 2015. 36(6): p. 395–405.

Khangulov, S.V., et al., L-arginine binding to liver arginase requires proton transfer to gateway residue His141 and coordination of the guanidinium group to the dimanganese (II, II) center. Biochemistry, 1998. 37(23): p. 8539–8550.

Romero, P.A., et al., SCHEMA-designed variants of human Arginase I and II reveal sequence elements important to stability and catalysis. ACS Synth Biol, 2012. 1(6): p. 221–8.

Stone, E.M., et al., Replacing Mn(2+) with Co(2+) in human arginase i enhances cytotoxicity toward l-arginine auxotrophic cancer cell lines. ACS Chem Biol, 2010. 5(3): p. 333–42.

Srivastava, S. and B.K. Ratha, Unique hepatic cytosolic arginase evolved independently in ureogenic freshwater air-breathing teleost, Heteropneustes fossilis. PLoS One, 2013. 8(6): p. e66057.

Ash, D.E., Structure and function of arginases. J Nutr, 2004. 134(10 Suppl): p. 2760S-2764S; discussion 2765S-2767S.

Lavulo, L.T., et al., Subunit-subunit interactions in trimeric arginase. Generation of active monomers by mutation of a single amino acid. J Biol Chem, 2001. 276(17): p. 14242–8.

Long, Y., et al., Argininosuccinate synthetase 1 (ASS1) is a common metabolic marker of chemosensitivity for targeted arginine- and glutamine-starvation therapy. Cancer Lett, 2017. 388: p. 54–63.

Cheng, N.M., Y.C. Leung, and W.H. Lo, Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation. 2014, Google Patents.

Cheng, P.N.-M., et al., Pegylated recombinant human arginase (rhArg-peg5, 000mw) inhibits the in vitro and in vivo proliferation of human hepatocellular carcinoma through arginine depletion. Cancer research, 2007. 67(1): p. 309–317.

Hsueh, E.C., et al., Deprivation of arginine by recombinant human arginase in prostate cancer cells. J Hematol Oncol, 2012. 5: p. 17.

Xiong, L., et al., Arginine Metabolism in Bacterial Pathogenesis and Cancer Therapy. Int J Mol Sci, 2016. 17(3): p. 363.

Yau, T., et al., Preliminary efficacy, safety, pharmacokinetics, pharmacodynamics and quality of life study of pegylated recombinant human arginase 1 in patients with advanced hepatocellular carcinoma. Invest New Drugs, 2015. 33(2): p. 496–504.

Georgiou, G. and E. Stone, Compositions of engineered human arginases and methods for treating cancer. 2013, Google Patents.

Georgiou, G. and E. Stone, Methods for purifying pegylated arginase. 2014, Google Patents.

Agrawal, V., et al., In-vivo evaluation of human recombinant Co-arginase against A375 melanoma xenografts. Melanoma Res, 2014. 24(6): p. 556–67.

Athenex, Athenex Announces U.S. FDA Allowance of Investigational New Drug Application for PT01 (Pegtomarginase). 2019.

Yu, K.-M., et al. Design, Engineering, and Characterization of a Novel Long-Acting (Pegylated) Single Isomer Human Arginase for Arginine-Depriving Anti-Cancer Treatment. in Journal of Clinical Oncology. 2019. AMER SOC CLINICAL ONCOLOGY 2318 MILL ROAD, STE 800, ALEXANDRIA, VA 22314 USA.

Fee, C.J. and J.M. Van Alstine, Purification of pegylated proteins. Methods Biochem Anal, 2011. 54: p. 339–62.

Haeckel, A., et al., XTEN as Biological Alternative to PEGylation Allows Complete Expression of a Protease-Activatable Killin-Based Cytostatic. PLoS One, 2016. 11(6): p. e0157193.

Langenheim, J.F. and W.Y. Chen, Improving the pharmacokinetics/ pharmacodynamics of prolactin, GH, and their antagonists by fusion to a synthetic albumin-binding peptide. J Endocrinol, 2009. 203(3): p. 375–87.

Li, L., et al., An Engineered Arginase FC Protein Inhibits Tumor Growth In VitroHuman Arginase I (Arg I)—A Potential Broad-spectrum Anti-cancer Agent Datta et al.and In Vivo. Evid Based Complement Alternat Med, 2013. 2013: p. 423129.

Kontermann, R.E., Strategies for extended serum half-life of protein therapeutics. Curr Opin Biotechnol, 2011. 22(6): p. 868–76.

Kintzing, J.R., M.V. Filsinger Interrante, and J.R. Cochran, Emerging Strategies for Developing Next-Generation Protein Therapeutics for Cancer Treatment. Trends Pharmacol Sci, 2016. 37(12): p. 993–1008.

Strohl, W.R., Fusion Proteins for Half-Life Extension of Biologics as a Strategy to Make Biobetters. BioDrugs, 2015. 29(4): p. 215–39.

Haeckel, A., et al., XTEN-annexin A5: XTEN allows complete expression of long-circulating protein-based imaging probes as recombinant alternative to PEGylation. J Nucl Med, 2014. 55(3): p. 508–14.

Aghaabdollahian, S., et al., Enhancing bioactivity, physicochemical, and pharmacokinetic properties of a nano-sized, anti-VEGFR2 Adnectin, through PASylation technology. Scientific reports, 2019. 9(1): p. 1–14.

Yeboah, A., et al., Elastin-like polypeptides: A strategic fusion partner for biologics. Biotechnol Bioeng, 2016. 113(8): p. 1617–27.

Schlapschy, M., et al., Fusion of a recombinant antibody fragment with a homo-amino-acid polymer: effects on biophysical properties and prolonged plasma half-life. Protein Engineering, Design & Selection, 2007. 20(6): p. 273–284.

Wang, B., et al., Production of a therapeutic protein by fusing it with two fragments of the carboxyl-terminal peptide of human chorionic gonadotropin β-subunit in Pichia pastoris. Biotechnology letters, 2016. 38(5): p. 801–807.

Iyengar, A.S., et al., Protein chimerization: a new frontier for engineering protein therapeutics with improved pharmacokinetics. Journal of Pharmacology and Experimental Therapeutics, 2019. 370(3): p. 703–714.

Kuo, T.T. and V.G. Aveson, Neonatal Fc receptor and IgG-based therapeutics. MAbs, 2011. 3(5): p. 422–30.

Sockolosky, J.T., M.R. Tiffany, and F.C. Szoka, Engineering neonatal Fc receptor-mediated recycling and transcytosis in recombinant proteins by short terminal peptide extensions. Proceedings of the National Academy of Sciences, 2012. 109(40): p. 16095–16100.

Levin, D., et al., Fc fusion as a platform technology: potential for modulating immunogenicity. Trends Biotechnol, 2015. 33(1): p. 27–34.

Rath, T., et al., Fc-fusion proteins and FcRn: structural insights for longer-lasting and more effective therapeutics. Crit Rev Biotechnol, 2015. 35(2): p. 235–54.

Dennis, M.S., et al., Albumin binding as a general strategy for improving the pharmacokinetics of proteins. J Biol Chem, 2002. 277(38): p. 35035–43.

Nilsen, J., et al., Human and mouse albumin bind their respective neonatal Fc receptors differently. Scientific reports, 2018. 8(1): p. 1–12.

Adabi, E., et al., Evaluation of an albumin-binding domain protein fused to recombinant human il-2 and its effects on the bioactivity and serum half-life of the cytokine. Iranian biomedical journal, 2017. 21(2): p. 77.

Hopp, J., et al., The effects of affinity and valency of an albumin-binding domain (ABD) on the half-life of a single-chain diabody-ABD fusion protein. Protein Engineering, Design & Selection, 2010. 23(11): p. 827–834.

Jacobs, S.A., et al., Fusion to a highly stable consensus albumin binding domain allows for tunable pharmacokinetics. Protein Eng Des Sel, 2015. 28(10): p. 385–93.

Sand, K.M.K., et al., Unraveling the interaction between FcRn and albumin: opportunities for design of albumin-based therapeutics. Frontiers in immunology, 2015. 5: p. 682.

Zhao, S., et al., Extending the serum half-life of G-CSF via fusion with the domain III of human serum albumin. Biomed Res Int, 2013. 2013: p. 107238.

Leung, Y.C., Composition and Application of Arginine-depleting Agents for Cancer, Obesity, Metabolic Disorders, and Related Complications and Comorbidities. 2020, Google Patents.

Wally, J., et al., The crystal structure of iron-free human serum transferrin provides insight into inter-lobe communication and receptor binding. J Biol Chem, 2006. 281(34): p. 24934–44.

Chen, X., J.L. Zaro, and W.C. Shen, Pharmacokinetics of recombinant bifunctional fusion proteins. Expert Opin Drug Metab Toxicol, 2012. 8(5): p. 581–95.

Qi, J., et al., Improving the specific antitumor efficacy of ONC by fusion with N-terminal domain of transferrin. Biosci Biotechnol Biochem, 2018. 82(7): p. 1153–1158.

Chen, X., J.L. Zaro, and W.C. Shen, Fusion protein linkers: property, design and functionality. Adv Drug Deliv Rev, 2013. 65(10): p. 1357–69.

Bai, Y., D.K. Ann, and W.-C. Shen, Recombinant granulocyte colony-stimulating factor-transferrin fusion protein as an oral myelopoietic agent. Proceedings of the National Academy of Sciences, 2005. 102(20): p. 7292–7296.




DOI: https://doi.org/10.37591/rrjooh.v9i3.2312

Refbacks

  • There are currently no refbacks.


Copyright (c) 2020 Research & Reviews: Journal of Oncology and Hematology