Unlocking the Potential of Arginine Deprivation Therapy: Recent Breakthroughs and Promising Future for Cancer Treatment
Abstract
:1. Introduction
2. Anticancer Treatments Using Arginine Deprivation Therapy
3. Enzyme-Mediated Arginine Deprivation Agents for Cancer Therapy: Pre-Clinical and Clinical Evidence
3.1. ADI
3.1.1. Preclinical Evidence Supporting ADI as a Potential Strategy for Cancer Therapy
3.1.2. Overview of Clinical Trials Using ADI-PEG 20 for Anticancer Therapy and Their Outcomes
Title | Status | Conditions | Interventions | Phases | Start Date | Enrollment | Outcome | NCT | Ref |
---|---|---|---|---|---|---|---|---|---|
ADI-PEG in Patients with Metastatic Melanoma | Completed | Melanoma | ADI-PEG 20 | Phase I | 2001/9/1 | 15 | NA | NCT00029900 | |
Testing of ADI-PEG in Hepatocellular Carcinoma | Completed | HCC | ADI-PEG 20 | Phase II | 2002/9/1 | 34 | NA | NCT00056992 | |
Pegylated Arginine Deiminase in Treating Patients with Metastatic Melanoma That Cannot Be Removed by Surgery | Completed | Melanoma | ADI-PEG 20 | Phase II | 2004/6/1 | 38 | 38 subjects were enrolled, 10 were ASS1 positive, and 17 were ASS1 negative. Median OS was 9.3 vs. 14.6 months for ASS1 positive vs. ASS1 negative, respectively. Median TTP was 1.8 vs. 3.6 months for ASS1 positive vs. ASS1 negative, respectively. 11 subjects who declined pre-treatment biopsies were not assessed. | NCT00450372 | [106] |
Study of ADI-PEG 20 in Patients with Advanced Melanoma | Completed | Melanoma | ADI PEG 20 | Phase I/II | 2007/7/1 | 31 | No objective responses were seen, but 9 patients had SD. Pharmacodynamic analysis showed complete plasma arginine depletion in 30 of 31 patients. | NCT00520299 | [107] |
Study of ADI-PEG 20 in Patients with Relapsed Sensitive or Refractory Small Cell Lung Cancer | Terminated | SCLC | ADI-PEG 20 | Phase II | 2011/1/1 | 22 | 20 evaluable patients were included in two cohorts: 8 with “sensitive” disease and 12 with “refractory” disease. 2 out of 8 had SD in the “sensitive” cohort and 2 out of 12 had SD in the “refractory” cohort. Pharmacodynamic analysis showed complete plasma arginine depletion in 21 out of 22 patients. | NCT01266018 | |
A Clinical Trial of ADI-PEG 20TM in Patients with Malignant Pleural Mesothelioma | Unknown | MPM | ADI-PEG 20 | Phase II | 2011/1/1 | 70 | 68 patients were evaluated, with a median PFS of 3.2 vs. 2.0 months (p = 0.03) in the ADI-PEG 20 group vs. best supportive care group. There were no significant differences between groups in response rate, OS, and adverse event rate. The best response observed was SD. | NCT01279967 | [111] |
Ph 3 ADI-PEG 20 Versus Placebo in Subjects with Advanced Hepatocellular Carcinoma Who Have Failed Prior Systemic Therapy | Completed | HCC | ADI-PEG 20 | Phase III | 2011/7/1 | 636 | 635 patients were evaluated, with 70% progressing on sorafenib. Median OS was 7.8 months for ADI-PEG 20 and 7.4 months for placebo (p = 0.88, HR = 1.02), while median PFS was 2.6 months for both (p = 0.07, HR = 1.17). The death rate within 30 days of the end of treatment was 15.2% on ADI-PEG 20 and 10.4% on placebo, with none related to therapy. A trend towards improved OS was observed in patients with more prolonged arginine depletion. | NCT01287585 | [103] |
Ph 1 Trial of ADI-PEG 20 Plus Docetaxel in Solid Tumors with Emphasis on Prostate Cancer and Non-Small Cell Lung Cancer | Completed | Solid Tumors and Prostate Cancer | ADI-PEG 20 | Phase I | 2011/9/6 | 43 | NA | NCT01497925 | |
Ph 1 Trial of ADI-PEG 20 Plus Cisplatin in Patients with Metastatic Melanoma | Completed | Melanoma and OC | ADI-PEG 20 | Phase I | 2012/9/1 | 8 | NA | NCT01665183 | |
PH 2 ADI-PEG 20 Study in Non-Hodgkin’s Lymphoma Subjects Who Have Failed Prior Systemic Therapy | Completed | NHL | ADI-PEG 20 | Phase II | 2013/12/6 | 18 | NA | NCT01910025 | |
Ph 1 ADI-PEG 20 Plus Doxorubicin; Patients with HER2 Negative Metastatic Breast Cancer | Completed | HER2(-ve) Breast Cancer | ADI-PEG 20 | Phase I | 2014/4/1 | 15 | NA | NCT01948843 | |
Ph 1 Study in Subjects with Tumors Requiring Arginine to Assess ADI-PEG 20 With Pemetrexed and Cisplatin | Terminated | MPM, NSCLC, Melanoma, HCC, Glioma, and Sarcoma | ADI-PEG 20 | Phase I | 2014/4/23 | 85 | Results were reported in 3 studies. In the first study, 7 patients had SD with a median PFS of 3.0 months and a median OS of 11.5 months. In the second study, the DCR was 85.7% with a PR rate of 47.6%. Median PFS and OS were 4.2 and 7.2 months, respectively. In the third study, treatment was well tolerated with a best overall response of SD in 8 patients. Median PFS was 5.2 months and OS was 6.3 months. | NCT02029690 | [92,96,110] |
Ph 2 Trial of ADI PEG 20 Plus Concurrent Transarterial Chemoembolization (TACE) vs. TACE Alone in Patients with Unresectable Hepatocellular Carcinoma | Completed | Unresectable HCC | ADI-PEG 20 + TACE | Phase II | 2014/10/15 | 30 | NA | NCT02006030 | |
Ph 1-2 Study ADI-PEG 20 Plus FOLFOX in Subjects with Advanced GI Malignancies Focusing on Hepatocellular Carcinoma | Terminated | Advanced GI cancer | ADI-PEG 20 + mFOLFOX6 | Phase I/II | 2014/11/1 | 140 | In the phase 1 trial, 27 patients were enrolled (23 with advanced HCC and 4 with other GI tumors). No dose-limiting toxicities were observed in cohort 1 or 2, and the recommended phase 2 dose for ADI-PEG 20 was 36 mg/m2 weekly with mFOLFOX6. The ORR was 21% (95% CI 7.5–43.7), with a median PFS and OS of 7.3 and 14.5 months, respectively. Arginine levels were depleted despite low levels of anti-ADI-PEG 20 antibodies, but there was no correlation between arginine depletion at 4 and 8 weeks and archival tumoral ASS1 levels with response | NCT02102022 | [102] |
Ph 1 Trial of ADI PEG 20 Plus Sorafenib to Treat Patients with Liver Cancer | Completed | HCC | ADI-PEG 20 | Phase I | 2014/11/1 | 8 | NA | NCT02101593 | |
Ph 1B Trial With ADI-PEG 20 Plus Nab-Paclitaxel and Gemcitabine in Subjects with Pancreatic Cancer | Completed | Pancreatic Cancer | ADI-PEG 20 | Phase I | 2014/11/17 | 21 | NA | NCT02101580 | |
PH 2 ADI-PEG 20 Acute Myeloid Leukemia | Completed | AML | ADI-PEG 20 | Phase II | 2015/1/6 | 43 | Among 21 evaluated patients, 2 (9.5%) achieved CR and 7 (33.3%) had stable disease, resulting in a DCR of 42.9%. The duration of response for the two CR patients was 7.5 and 8.8 months. DCR was associated with arginine depletion to ≤10 μM for a median of 8 weeks. | NCT01910012 | [113] |
Ph 1 Study of ADI-PEG 20 Plus Low Dose Cytarabine in Older Patients With AML | Terminated | AML | ADI-PEG 20 + Cytarabine | Phase I | 2017/1/20 | 23 | 23 patients were given low-dose cytarabine subcutaneously twice daily for 10 days every 28 days and ADI-PEG20 at 18 or 36 mg/m2 (dose levels 1 and 2) intramuscularly weekly. 18 evaluable patients had a 44.4% ORR and median OS of 8.0 (4.5-not reached) months. In 7 treatment-naïve patients, the ORR was 71.4% with 57.1% complete remission rate. No dose-limiting toxicities were reported, and the combination was well-tolerated. | NCT02875093 | [114] |
Study ADI-PEG 20 Plus Pembrolizumab in Advanced Solid Cancers | Terminated | Advanced Solid Cancers | ADI PEG 20 + Pembrolizumab | Phase I | 2017/7/14 | 33 | 25 patients were evaluated; 9 in the dose-escalation cohort and 16 in the expansion cohort. Mean arginine levels were suppressed for 1–3 weeks but gradually increased. CD3+ T cells increased in 10 of 12 subjects (83.3%) after ADI-PEG 20 treatment, including 3 PR (p = 0.02). PD-L1 expression was low and increased in 3 of 10 subjects (30%). 6 of 25 heavily pretreated patients had PR, including both ASS1 proficient and deficient subjects, for a response rate of 24%. | NCT03254732 | [117] |
Ph 2/3 Study in Subjects with MPM to Assess ADI-PEG 20 With Pemetrexed and Cisplatin | Completed | MPM | ADI-PEG 20 + Pemetrexed + Cisplatin | Phase II/III | 2017/8/1 | 249 | The plasma arginine decreased while citrulline increased and was maintained over 18 weeks of ADIPemCis therapy. In 31 pilot evaluable patients, the DCR was 93.5% (n = 29/31; 95% CI: 78.6–99.2%), with a PR rate of 35.5% (n = 11/31; 95% CI: 19.2–54.6%). The median PFS and OS were 5.6 (4.0–6.0) and 10.1 (6.1–11.1) months, respectively. The treatment was well tolerated. | NCT02709512 | [112,118] |
ADI-PEG 20 in Combination with Gemcitabine and Docetaxel for the Treatment of Soft Tissue Sarcoma, Osteosarcoma, Ewing’s Sarcoma, and Small Cell Lung Cancer | Completed | Sarcoma | ADI-PEG 20 + Gemcitabine + Docetaxel | Phase II | 2018/5/9 | 98 | 75 patients were evaluated with PFS/OS (months) results of 6.0/N.D. for the 600 mg/m2 group (n = 31), 7.2/22.5 for leiomyosarcoma (LMS) (n = 33), 5.1/17.4 for liposarcoma, and 2.8/15.0 for other (n = 36). Out of 75 patients, 8% (6/75) had CR (3 LMS, 1 synovial, and 2 angiosarcoma), 17% (13/75) had PR, and 43% (32/75) had SD, leading to an ORR of 25% (19/75) and a clinical benefit rate of 68% (51/75). There was a tendency for ASS1 negative tumors to benefit more than ASS1 positive tumors. | NCT03449901 | [119] |
Study in Patients With Tumours Requiring Arginine to Assess ADI-PEG 20 With Atezolizumab, Pemetrexed and Carboplatin | Withdrawn | NSCLC | Atezolizumab + Pemetrexed + Carboplatin + ADI PEG 20 | Phase I | 2018/6/1 | 0 | NA | NCT03498222 | |
Study of Immunotherapy Plus ADI-PEG 20 for the Treatment of Advanced Uveal Melanoma | Completed | Melanoma | ADI PEG 20 + Nivolumab + Ipilimumab | Phase I | 2019/4/16 | 9 | NA | NCT03922880 | |
ADI-PEG 20 Plus Radiotherapy and Temozolomide in Subjects with Glioblastoma Multiforme | Recruiting | GBM | ADI-PEG 20 + Temozolomide | Phase I | 2020/9/14 | Estimated 32 | NA | NCT04587830 | |
Study of ADI-PEG 20 Versus Placebo in Subjects with Genotype WWOX-GG, Unresectable HCC | Recruiting | HCC | ADI-PEG 20 | Phase III | 2022/3/14 | Estimated 150 | NA | NCT05317819 | |
Study of ADI-PEG 20, Venetoclax and Azacitidine in Acute Myeloid Leukemia | Recruiting | AML | ADI-PEG 20 | Phase I | 2022/4/5 | Estimated 60 | NA | NCT05001828 | |
Nivolumab and ADI-PEG 20 Before Surgery for the Treatment of Resectable Liver Cancer | Withdrawn | Resectable HCC | Nivolumab + ADI-PEG 20 + Resection | Phase II | 2022/4/13 | 0 | NA | NCT04965714 | |
ADI-PEG 20 in Combination with Gemcitabine and Docetaxel After Progression on Frontline Therapy in Non-small Cell and Small Cell Lung Cancers | Not yet recruiting | NSCLC and SCLC | ADI-PEG 20 + Gemcitabine + Docetaxel | Phase I/II | 2023/3/31 | Estimated 108 | NA | NCT05616624 | |
Study of ADI-PEG 20 or Placebo Plus Gem and Doc in Previously Treated Subjects with Leiomyosarcoma (ARGSARC) | Not yet recruiting | Sarcoma | ADI PEG 20 | Phase III | 2023/6/15 | Estimated 300 | NA | NCT05712694 |
3.2. rhArg1
3.2.1. Preclinical Evidence Supporting rhArg1 as a Potential Strategy for Anticancer Therapy
3.2.2. Overview of Clinical Trials Using rhArg1-PEG for Cancer Therapy and Their Outcomes
Title | Status | Conditions | Interventions | Phases | Start Date | Enrollment | Outcome | NCT | Ref |
---|---|---|---|---|---|---|---|---|---|
Study of PEGylated Human Recombinant Arginase for Liver Cancer | Completed | HCC | rhArg1-PEG + Doxorubicin | Phase I | 2008/5/1 | 15 | NA | NCT00988195 | |
Study of PEGylated Human Recombinant Arginase for Liver Cancer (BCT-100-002) | Completed | HCC | rhArg1-PEG | Phase I/II | 2010/3/1 | 20 | May be included in NCT02089763. | NCT01092091 | [152] |
A Pilot Study of Recombinant Human Arginase 1 (rhArg1) in Patients with Relapsed or Refractory Leukemia or Lymphoma | Terminated | Leukemia and Lymphoma | rhArg1-PEG | Phase I | 2012/4/1 | 1 | NA | NCT01551628 | |
Efficacy Study of PEGylated Recombinant Human Arginase 1 as a Second-line Therapy in Patients with Advanced Liver Cancer | Completed | HCC | rhArg1-PEG | Phase II | 2014/4/1 | 27 | 27 patients were recruited, with a median TTP and PFS of 6 (95% CI, 5.9–6.0) weeks and a DCR of 21.7% (5 SD). The drug was well tolerated. Duration of arginine depletion correlated with OS. Among patients with available IHC results, those with ASS1-negative tumors had an OS of 35 (95% CI: 8.3–78.0) weeks, compared to 15.14 (95% CI: 13.4–15.1) weeks in those with ASS1-positive tumors. | NCT02089763 | [152] |
PEGylated Recombinant Human Arginase 1 in Combination with Oxaliplatin and Capecitabine for the Treatment of HCC | Completed | HCC | rhArg1-PEG + Oxaliplatin + Capecitabine | Phase II | 2014/4/1 | 17 | 17 patients received oxaliplatin at 3 dose levels: 85 mg/m2 (8 patients), 100 mg/m2 (3 patients), and 130 mg/m2 (6 patients), with no dose-limiting toxicity. Median study duration was 8 weeks. Among 14 evaluable cases, one achieved PR, 4 had SD, and the DCR was 36%. Most responses occurred in the 130 mg/m2 cohort with 1 PR and 2 SD. Median TTP and PFS were both 7.0 weeks. Overall median OS was 10.7 months, and median OS was not reached at 19.4 months of follow-up in the 130 mg/m2 cohort. | NCT02089633 | [153] |
Recombinant Human Arginase 1 (rhArg1) in Patients with Advanced Arginine Auxotrophic Solid Tumors | Completed | Melanoma and Prostate cancer | rhArg1-PEG | Phase I | 2014/11/1 | 23 | A 65-year-old patient with metastatic melanoma, who failed two immunotherapy strategies, received 2 mg/kg intravenously weekly BCT-100. The patient had no toxicities > grade 2 and achieved CR for over 30 months. The tumor lacked expression of ASS1 and OTC. | NCT02285101 | [155] |
Efficacy and Safety Study of Recombinant Human Arginase 1 in Patients with Relapsed or Refractory Acute Myeloid Leukemia | Unknown | AML | rhArg1-PEG | Phase II | 2016/9/1 | Estimated 25 | NA | NCT02899286 | |
A Study Evaluating the Safety and Activity of PEGylated Recombinant Human Arginase (BCT-100) | Completed | AML and ALL | rhArg1-PEG | Phase I/II | 2018/8/28 | 49 | NA | NCT03455140 |
3.3. rhADC
3.4. BCA
4. Potential Strategies for Arginine Deprivation Therapy in Cancer Independent of Arginine-Depleting Enzymes
5. Biomarkers for Arginine Deprivation Therapy in Cancers
5.1. Tissue protein Level
5.1.1. ASS1
5.1.2. HIF-1
5.1.3. WWOX
5.2. Genetic Biomarker
5.3. Plasma Biomarker
6. Conclusions and Future Perspective
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Sivanand, S.; Vander Heiden, M.G. Emerging Roles for Branched-Chain Amino Acid Metabolism in Cancer. Cancer Cell 2020, 37, 147–156. [Google Scholar] [CrossRef]
- Vettore, L.; Westbrook, R.L.; Tennant, D.A. New aspects of amino acid metabolism in cancer. Br. J. Cancer 2020, 122, 150–156. [Google Scholar] [CrossRef]
- Wu, G. Functional amino acids in growth, reproduction, and health. Adv. Nutr. 2010, 1, 31–37. [Google Scholar] [CrossRef] [Green Version]
- Tennant, D.A.; Duran, R.V.; Gottlieb, E. Targeting metabolic transformation for cancer therapy. Nat. Rev. Cancer 2010, 10, 267–277. [Google Scholar] [CrossRef]
- Lieu, E.L.; Nguyen, T.; Rhyne, S.; Kim, J. Amino acids in cancer. Exp. Mol. Med. 2020, 52, 15–30. [Google Scholar] [CrossRef]
- Pokrovsky, V.S.; Abo Qoura, L.; Morozova, E.; Bunik, V.I. Predictive markers for efficiency of the amino-acid deprivation therapies in cancer. Front. Med. 2022, 9, 1035356. [Google Scholar] [CrossRef]
- Kumar, R.; Mishra, A.; Gautam, P.; Feroz, Z.; Vijayaraghavalu, S.; Likos, E.M.; Shukla, G.C.; Kumar, M. Metabolic Pathways, Enzymes, and Metabolites: Opportunities in Cancer Therapy. Cancers 2022, 14, 5268. [Google Scholar] [CrossRef]
- Patil, M.D.; Bhaumik, J.; Babykutty, S.; Banerjee, U.C.; Fukumura, D. Arginine dependence of tumor cells: Targeting a chink in cancer’s armor. Oncogene 2016, 35, 4957–4972. [Google Scholar] [CrossRef]
- Shosha, E.; Fouda, A.Y.; Narayanan, S.P.; Caldwell, R.W.; Caldwell, R.B. Is the Arginase Pathway a Novel Therapeutic Avenue for Diabetic Retinopathy? J. Clin. Med. 2020, 9, 425. [Google Scholar] [CrossRef] [Green Version]
- Morettin, A.; Baldwin, R.M.; Cote, J. Arginine methyltransferases as novel therapeutic targets for breast cancer. Mutagenesis 2015, 30, 177–189. [Google Scholar] [CrossRef] [Green Version]
- Rath, M.; Muller, I.; Kropf, P.; Closs, E.I.; Munder, M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front. Immunol. 2014, 5, 532. [Google Scholar] [CrossRef] [Green Version]
- Morris, S.M., Jr. Arginine Metabolism Revisited. J. Nutr. 2016, 146, 2579S–2586S. [Google Scholar] [CrossRef] [Green Version]
- Keshet, R.; Szlosarek, P.; Carracedo, A.; Erez, A. Rewiring urea cycle metabolism in cancer to support anabolism. Nat. Rev. Cancer 2018, 18, 634–645. [Google Scholar] [CrossRef]
- Marti, I.L.A.A.; Reith, W. Arginine-dependent immune responses. Cell. Mol. Life Sci. 2021, 78, 5303–5324. [Google Scholar] [CrossRef]
- Morris, S.M., Jr. Arginine: Beyond protein. Am. J. Clin. Nutr. 2006, 83, 508S–512S. [Google Scholar] [CrossRef] [Green Version]
- Appleton, J. Arginine: Clinical potential of a semi-essential amino acid. Altern. Med. Rev. 2002, 7, 512–522. [Google Scholar]
- Chen, C.L.; Hsu, S.C.; Ann, D.K.; Yen, Y.; Kung, H.J. Arginine Signaling and Cancer Metabolism. Cancers 2021, 13, 3541. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Watford, M. The urea cycle: Teaching intermediary metabolism in a physiological setting. Biochem. Mol. Biol. Educ. 2003, 31, 289–297. [Google Scholar] [CrossRef]
- Dillon, B.J.; Prieto, V.G.; Curley, S.A.; Ensor, C.M.; Holtsberg, F.W.; Bomalaski, J.S.; Clark, M.A. Incidence and distribution of argininosuccinate synthetase deficiency in human cancers: A method for identifying cancers sensitive to arginine deprivation. Cancer 2004, 100, 826–833. [Google Scholar] [CrossRef]
- Delage, B.; Fennell, D.A.; Nicholson, L.; McNeish, I.; Lemoine, N.R.; Crook, T.; Szlosarek, P.W. Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. Int. J. Cancer 2010, 126, 2762–2772. [Google Scholar] [CrossRef]
- Haines, R.J.; Pendleton, L.C.; Eichler, D.C. Argininosuccinate synthase: At the center of arginine metabolism. Int. J. Biochem. Mol. Biol. 2011, 2, 8–23. [Google Scholar]
- Choy, C.T.; Wong, C.H.; Loong, H.H.F. Low expressions of ASS1 and OTC in glioblastoma suggest the potential clinical use of recombinant human arginase (rhArg). J. Neuro-Oncol. 2016, 129, 579–581. [Google Scholar] [CrossRef]
- Miao, Y.Q.; Chen, W.; Zhou, J.; Shen, Q.; Sun, Y.; Li, T.; Wang, S.C. N(6)-adenosine-methyltransferase-14 promotes glioma tumorigenesis by repressing argininosuccinate synthase 1 expression in an m6A-dependent manner. Bioengineered 2022, 13, 1858–1871. [Google Scholar] [CrossRef]
- Giatromanolaki, A.; Harris, A.L.; Koukourakis, M.I. The prognostic and therapeutic implications of distinct patterns of argininosuccinate synthase 1 (ASS1) and arginase-2 (ARG2) expression by cancer cells and tumor stroma in non-small-cell lung cancer. Cancer Metab. 2021, 9, 28. [Google Scholar] [CrossRef]
- Walts, A.E.; Bomalaski, J.S.; Ines, D.; Orsulic, S. Argininosuccinate synthetase (ASS) deficiency in high-grade pulmonary neuroendocrine carcinoma: An opportunity for personalized targeted therapy. J. Cancer Res. Clin. Oncol. 2015, 141, 1363–1369. [Google Scholar] [CrossRef]
- Huang, C.C.; Tsai, S.T.; Kuo, C.C.; Chang, J.S.; Jin, Y.T.; Chang, J.Y.; Hsiao, J.R. Arginine deprivation as a new treatment strategy for head and neck cancer. Oral Oncol. 2012, 48, 1227–1235. [Google Scholar] [CrossRef]
- Chu, Y.D.; Liu, H.F.; Chen, Y.C.; Chou, C.H.; Yeh, C.T. WWOX-rs13338697 genotype predicts therapeutic efficacy of ADI-PEG 20 for patients with advanced hepatocellular carcinoma. Front. Oncol. 2022, 12, 996820. [Google Scholar] [CrossRef]
- Liu, Q.; Stewart, J.; Wang, H.; Rashid, A.; Zhao, J.; Katz, M.H.; Lee, J.E.; Fleming, J.B.; Maitra, A.; Wolff, R.A.; et al. Reduced expression of argininosuccinate synthetase 1 has a negative prognostic impact in patients with pancreatic ductal adenocarcinoma. PLoS ONE 2017, 12, e0171985. [Google Scholar] [CrossRef] [Green Version]
- Yoon, C.Y.; Shim, Y.J.; Kim, E.H.; Lee, J.H.; Won, N.H.; Kim, J.H.; Park, I.S.; Yoon, D.K.; Min, B.H. Renal cell carcinoma does not express argininosuccinate synthetase and is highly sensitive to arginine deprivation via arginine deiminase. Int. J. Cancer 2007, 120, 897–905. [Google Scholar] [CrossRef]
- Sahu, D.; Gupta, S.; Hau, A.M.; Nakashima, K.; Leivo, M.Z.; Searles, S.C.; Elson, P.; Bomalaski, J.S.; Casteel, D.E.; Boss, G.R.; et al. Argininosuccinate Synthetase 1 Loss in Invasive Bladder Cancer Regulates Survival through General Control Nonderepressible 2 Kinase-Mediated Eukaryotic Initiation Factor 2alpha Activity and Is Targetable by Pegylated Arginine Deiminase. Am. J. Pathol. 2017, 187, 200–213. [Google Scholar] [CrossRef] [Green Version]
- Allen, M.D.; Luong, P.; Hudson, C.; Leyton, J.; Delage, B.; Ghazaly, E.; Cutts, R.; Yuan, M.; Syed, N.; Lo Nigro, C.; et al. Prognostic and therapeutic impact of argininosuccinate synthetase 1 control in bladder cancer as monitored longitudinally by PET imaging. Cancer Res. 2014, 74, 896–907. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; He, W.; Zhang, S. Seeking for Correlative Genes and Signaling Pathways With Bone Metastasis From Breast Cancer by Integrated Analysis. Front. Oncol. 2019, 9, 138. [Google Scholar] [CrossRef] [Green Version]
- Huang, H.Y.; Wu, W.R.; Wang, Y.H.; Wang, J.W.; Fang, F.M.; Tsai, J.W.; Li, S.H.; Hung, H.C.; Yu, S.C.; Lan, J.; et al. ASS1 as a novel tumor suppressor gene in myxofibrosarcomas: Aberrant loss via epigenetic DNA methylation confers aggressive phenotypes, negative prognostic impact, and therapeutic relevance. Clin. Cancer Res. 2013, 19, 2861–2872. [Google Scholar] [CrossRef] [Green Version]
- Ji, J.X.; Cochrane, D.R.; Tessier-Cloutier, B.; Chen, S.Y.; Ho, G.; Pathak, K.V.; Alcazar, I.N.; Farnell, D.; Leung, S.; Cheng, A.; et al. Arginine Depletion Therapy with ADI-PEG20 Limits Tumor Growth in Argininosuccinate Synthase-Deficient Ovarian Cancer, Including Small-Cell Carcinoma of the Ovary, Hypercalcemic Type. Clin. Cancer Res. 2020, 26, 4402–4413. [Google Scholar] [CrossRef]
- Delage, B.; Luong, P.; Maharaj, L.; O’Riain, C.; Syed, N.; Crook, T.; Hatzimichael, E.; Papoudou-Bai, A.; Mitchell, T.J.; Whittaker, S.J.; et al. Promoter methylation of argininosuccinate synthetase-1 sensitises lymphomas to arginine deiminase treatment, autophagy and caspase-dependent apoptosis. Cell Death Dis. 2012, 3, e342. [Google Scholar] [CrossRef] [Green Version]
- Miraki-Moud, F.; Ghazaly, E.; Ariza-McNaughton, L.; Hodby, K.A.; Clear, A.; Anjos-Afonso, F.; Liapis, K.; Grantham, M.; Sohrabi, F.; Cavenagh, J.; et al. Arginine deprivation using pegylated arginine deiminase has activity against primary acute myeloid leukemia cells in vivo. Blood 2015, 125, 4060–4068. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Chung, S.F.; Tam, S.Y.; Leung, Y.C.; Guan, X. Arginine deprivation as a strategy for cancer therapy: An insight into drug design and drug combination. Cancer Lett. 2021, 502, 58–70. [Google Scholar] [CrossRef]
- Veronese, F.M.; Pasut, G. PEGylation, successful approach to drug delivery. Drug Discov. Today 2005, 10, 1451–1458. [Google Scholar] [CrossRef]
- Roopenian, D.C.; Akilesh, S. FcRn: The neonatal Fc receptor comes of age. Nat. Rev. Immunol. 2007, 7, 715–725. [Google Scholar] [CrossRef]
- Shirai, H.; Blundell, T.L.; Mizuguchi, K. A novel superfamily of enzymes that catalyze the modification of guanidino groups. Trends Biochem. Sci. 2001, 26, 465–468. [Google Scholar] [CrossRef]
- Kondo, K.; Sone, H.; Yoshida, H.; Toida, T.; Kanatani, K.; Hong, Y.-M.; Nishino, N.; Tanaka, J.-i. Cloning and sequence analysis of the arginine deiminase gene from Mycoplasma arginini. Mol. Gen. Genet. 1990, 221, 81–86. [Google Scholar] [CrossRef]
- Ohno, T.; Ando, O.; Sugimura, K.; Taniai, M.; Suzuki, M.; Fukuda, S.; Nagase, Y.; Yamamoto, K.; Azuma, I. Cloning and nucleotide sequence of the gene encoding arginine deiminase of Mycoplasma arginini. Infect. Immun. 1990, 58, 3788–3795. [Google Scholar] [CrossRef] [Green Version]
- Weickmann, J.L.; Fahrney, D.E. Arginine deiminase from Mycoplasma arthritidis. Evidence for multiple forms. J. Biol. Chem. 1977, 252, 2615–2620. [Google Scholar] [CrossRef]
- Kelly, M.P.; Jungbluth, A.A.; Wu, B.W.; Bomalaski, J.; Old, L.J.; Ritter, G. Arginine deiminase PEG20 inhibits growth of small cell lung cancers lacking expression of argininosuccinate synthetase. Br. J. Cancer 2012, 106, 324–332. [Google Scholar] [CrossRef] [Green Version]
- Shen, L.-J.; Shen, W.-C. Drug evaluation: ADI-PEG-20--a PEGylated arginine deiminase for arginine-auxotrophic cancers. Curr. Opin. Mol. Ther. 2006, 8, 240–248. [Google Scholar]
- Syed, N.; Langer, J.; Janczar, K.; Singh, P.; Lo Nigro, C.; Lattanzio, L.; Coley, H.M.; Hatzimichael, E.; Bomalaski, J.; Szlosarek, P.; et al. Epigenetic status of argininosuccinate synthetase and argininosuccinate lyase modulates autophagy and cell death in glioblastoma. Cell Death Dis. 2013, 4, e458. [Google Scholar] [CrossRef] [Green Version]
- Hajji, N.; Garcia-Revilla, J.; Soto, M.S.; Perryman, R.; Symington, J.; Quarles, C.C.; Healey, D.R.; Guo, Y.; Orta-Vazquez, M.L.; Mateos-Cordero, S.; et al. Arginine deprivation alters microglial polarity and synergizes with radiation to eradicate non-arginine-auxotrophic glioblastoma tumors. J. Clin. Investig. 2022, 132, e142137. [Google Scholar] [CrossRef]
- Moren, L.; Perryman, R.; Crook, T.; Langer, J.K.; Oneill, K.; Syed, N.; Antti, H. Metabolomic profiling identifies distinct phenotypes for ASS1 positive and negative GBM. BMC Cancer 2018, 18, 167. [Google Scholar] [CrossRef] [Green Version]
- Przystal, J.M.; Hajji, N.; Khozoie, C.; Renziehausen, A.; Zeng, Q.; Abaitua, F.; Hajitou, A.; Suwan, K.; Want, E.; Bomalaski, J.; et al. Efficacy of arginine depletion by ADI-PEG20 in an intracranial model of GBM. Cell Death Dis. 2018, 9, 1192. [Google Scholar] [CrossRef] [Green Version]
- Ensor, C.M.; Holtsberg, F.W.; Bomalaski, J.S.; Clark, M.A. Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 2002, 62, 5443–5450. [Google Scholar]
- McAlpine, J.A.; Lu, H.-T.; Wu, K.C.; Knowles, S.K.; Thomson, J.A. Down-regulation of argininosuccinate synthetase is associated with cisplatin resistance in hepatocellular carcinoma cell lines: Implications for PEGylated arginine deiminase combination therapy. BMC Cancer 2014, 14, 621. [Google Scholar] [CrossRef] [Green Version]
- Thongkum, A.; Wu, C.; Li, Y.Y.; Wangpaichitr, M.; Navasumrit, P.; Parnlob, V.; Sricharunrat, T.; Bhudhisawasdi, V.; Ruchirawat, M.; Savaraj, N. The Combination of Arginine Deprivation and 5-Fluorouracil Improves Therapeutic Efficacy in Argininosuccinate Synthetase Negative Hepatocellular Carcinoma. Int. J. Mol. Sci. 2017, 18, 1175. [Google Scholar] [CrossRef] [Green Version]
- Bowles, T.L.; Kim, R.; Galante, J.; Parsons, C.M.; Virudachalam, S.; Kung, H.J.; Bold, R.J. Pancreatic cancer cell lines deficient in argininosuccinate synthetase are sensitive to arginine deprivation by arginine deiminase. Int. J. Cancer 2008, 123, 1950–1955. [Google Scholar] [CrossRef] [Green Version]
- Daylami, R.; Muilenburg, D.J.; Virudachalam, S.; Bold, R.J. Pegylated arginine deiminase synergistically increases the cytotoxicity of gemcitabine in human pancreatic cancer. J. Exp. Clin. Cancer Res. 2014, 33, 102. [Google Scholar] [CrossRef]
- Prudner, B.C.; Rathore, R.; Robinson, A.M.; Godec, A.; Chang, S.F.; Hawkins, W.G.; Hirbe, A.C.; Van Tine, B.A. Arginine Starvation and Docetaxel Induce c-Myc-Driven hENT1 Surface Expression to Overcome Gemcitabine Resistance in ASS1-Negative Tumors. Clin. Cancer Res. 2019, 25, 5122–5134. [Google Scholar] [CrossRef] [Green Version]
- Singh, P.K.; Deorukhkar, A.A.; Venkatesulu, B.P.; Li, X.; Tailor, R.; Bomalaski, J.S.; Krishnan, S. Exploiting Arginine Auxotrophy with Pegylated Arginine Deiminase (ADI-PEG20) to Sensitize Pancreatic Cancer to Radiotherapy via Metabolic Dysregulation. Mol. Cancer Ther. 2019, 18, 2381–2393. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.S.; Xu, S.; Cui, J.; Poddar, S.; Le, T.M.; Hayrapetyan, H.; Li, L.; Wu, N.; Moore, A.M.; Zhou, L.; et al. Histone deacetylase inhibition is synthetically lethal with arginine deprivation in pancreatic cancers with low argininosuccinate synthetase 1 expression. Theranostics 2020, 10, 829–840. [Google Scholar] [CrossRef]
- Tu, M.J.; Duan, Z.; Liu, Z.; Zhang, C.; Bold, R.J.; Gonzalez, F.J.; Kim, E.J.; Yu, A.M. MicroRNA-1291-5p Sensitizes Pancreatic Carcinoma Cells to Arginine Deprivation and Chemotherapy through the Regulation of Arginolysis and Glycolysis. Mol. Pharmacol. 2020, 98, 686–694. [Google Scholar] [CrossRef]
- Kim, R.H.; Coates, J.M.; Bowles, T.L.; McNerney, G.P.; Sutcliffe, J.; Jung, J.U.; Gandour-Edwards, R.; Chuang, F.Y.; Bold, R.J.; Kung, H.J. Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res. 2009, 69, 700–708. [Google Scholar] [CrossRef] [Green Version]
- Kim, R.H.; Bold, R.J.; Kung, H.-J. ADI, autophagy and apoptosis: Metabolic stress as a therapeutic option for prostate cancer. Autophagy 2009, 5, 567–568. [Google Scholar] [CrossRef] [Green Version]
- Changou, C.A.; Chen, Y.R.; Xing, L.; Yen, Y.; Chuang, F.Y.; Cheng, R.H.; Bold, R.J.; Ann, D.K.; Kung, H.J. Arginine starvation-associated atypical cellular death involves mitochondrial dysfunction, nuclear DNA leakage, and chromatin autophagy. Proc. Natl. Acad. Sci. USA 2014, 111, 14147–14152. [Google Scholar] [CrossRef] [Green Version]
- Chu, C.Y.; Lee, Y.C.; Hsieh, C.H.; Yeh, C.T.; Chao, T.Y.; Chen, P.H.; Lin, I.H.; Hsieh, T.H.; Shih, J.W.; Cheng, C.H.; et al. Genome-wide CRISPR/Cas9 knockout screening uncovers a novel inflammatory pathway critical for resistance to arginine-deprivation therapy. Theranostics 2021, 11, 3624–3641. [Google Scholar] [CrossRef]
- Qiu, F.; Chen, Y.R.; Liu, X.; Chu, C.Y.; Shen, L.J.; Xu, J.; Gaur, S.; Forman, H.J.; Zhang, H.; Zheng, S.; et al. Arginine starvation impairs mitochondrial respiratory function in ASS1-deficient breast cancer cells. Sci. Signal 2014, 7, ra31. [Google Scholar] [CrossRef] [Green Version]
- Yeh, T.H.; Chen, Y.R.; Chen, S.Y.; Shen, W.C.; Ann, D.K.; Zaro, J.L.; Shen, L.J. Selective Intracellular Delivery of Recombinant Arginine Deiminase (ADI) Using pH-Sensitive Cell Penetrating Peptides To Overcome ADI Resistance in Hypoxic Breast Cancer Cells. Mol. Pharm. 2016, 13, 262–271. [Google Scholar] [CrossRef] [Green Version]
- Bean, G.R.; Kremer, J.C.; Prudner, B.C.; Schenone, A.D.; Yao, J.C.; Schultze, M.B.; Chen, D.Y.; Tanas, M.R.; Adkins, D.R.; Bomalaski, J.; et al. A metabolic synthetic lethal strategy with arginine deprivation and chloroquine leads to cell death in ASS1-deficient sarcomas. Cell Death Dis. 2016, 7, e2406. [Google Scholar] [CrossRef] [Green Version]
- Kremer, J.C.; Prudner, B.C.; Lange, S.E.S.; Bean, G.R.; Schultze, M.B.; Brashears, C.B.; Radyk, M.D.; Redlich, N.; Tzeng, S.C.; Kami, K.; et al. Arginine Deprivation Inhibits the Warburg Effect and Upregulates Glutamine Anaplerosis and Serine Biosynthesis in ASS1-Deficient Cancers. Cell Rep. 2017, 18, 991–1004. [Google Scholar] [CrossRef] [Green Version]
- Prudner, B.C.; Sun, F.; Kremer, J.C.; Xu, J.; Huang, C.; Sai, K.K.S.; Morgan, Z.; Leeds, H.; McConathy, J.; Van Tine, B.A. Amino Acid Uptake Measured by [18F]AFETP Increases in Response to Arginine Starvation in ASS1-Deficient Sarcomas. Theranostics 2018, 8, 2107–2116. [Google Scholar] [CrossRef]
- Rogers, L.C.; Zhou, J.; Baker, A.; Schutt, C.R.; Panda, P.K.; Van Tine, B.A. Intracellular arginine-dependent translation sensor reveals the dynamics of arginine starvation response and resistance in ASS1-negative cells. Cancer Metab. 2021, 9, 4. [Google Scholar] [CrossRef]
- Cheon, D.J.; Walts, A.E.; Beach, J.A.; Lester, J.; Bomalaski, J.S.; Walsh, C.S.; Ruprecht Wiedemeyer, W.; Karlan, B.Y.; Orsulic, S. Differential expression of argininosuccinate synthetase in serous and non-serous ovarian carcinomas. J. Pathol. Clin. Res. 2015, 1, 41–53. [Google Scholar] [CrossRef] [Green Version]
- Savaraj, N.; Wu, C.; Kuo, M.T.; You, M.; Wangpaichitr, M.; Robles, C.; Spector, S.; Feun, L. The relationship of arginine deprivation, argininosuccinate synthetase and cell death in melanoma. Drug Target Insights 2007, 2, 119–128. [Google Scholar] [CrossRef] [Green Version]
- Tsai, W.B.; Aiba, I.; Lee, S.Y.; Feun, L.; Savaraj, N.; Kuo, M.T. Resistance to arginine deiminase treatment in melanoma cells is associated with induced argininosuccinate synthetase expression involving c-Myc/HIF-1alpha/Sp4. Mol. Cancer Ther. 2009, 8, 3223–3233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- You, M.; Savaraj, N.; Wangpaichitr, M.; Wu, C.; Kuo, M.T.; Varona-Santos, J.; Nguyen, D.M.; Feun, L. The combination of ADI-PEG20 and TRAIL effectively increases cell death in melanoma cell lines. Biochem. Biophys. Res. Commun. 2010, 394, 760–766. [Google Scholar] [CrossRef] [Green Version]
- You, M.; Savaraj, N.; Kuo, M.T.; Wangpaichitr, M.; Varona-Santos, J.; Wu, C.; Nguyen, D.M.; Feun, L. TRAIL induces autophagic protein cleavage through caspase activation in melanoma cell lines under arginine deprivation. Mol. Cell. Biochem. 2013, 374, 181–190. [Google Scholar] [CrossRef] [Green Version]
- Stelter, L.; Evans, M.J.; Jungbluth, A.A.; Zanzonico, P.; Ritter, G.; Ku, T.; Rosenfeld, E.; Bomalaski, J.S.; Old, L.; Larson, S.M. Novel mechanistic insights into arginine deiminase pharmacology suggest 18F-FDG is not suitable to evaluate clinical response in melanoma. J. Nucl. Med. 2012, 53, 281–286. [Google Scholar] [CrossRef] [Green Version]
- Stelter, L.; Fuchs, S.; Jungbluth, A.A.; Ritter, G.; Longo, V.A.; Zanzonico, P.; Raschzok, N.; Sauer, I.M.; Bomalaski, J.S.; Larson, S.M. Evaluation of arginine deiminase treatment in melanoma xenografts using (18)F-FLT PET. Mol. Imaging Biol. 2013, 15, 768–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stelter, L.; Evans, M.J.; Jungbluth, A.A.; Longo, V.A.; Zanzonico, P.; Ritter, G.; Bomalaski, J.S.; Old, L.; Larson, S.M. Imaging of tumor vascularization using fluorescence molecular tomography to monitor arginine deiminase treatment in melanoma. Mol. Imaging 2013, 12, 67–73. [Google Scholar]
- Savaraj, N.; Wu, C.; Li, Y.Y.; Wangpaichitr, M.; You, M.; Bomalaski, J.; He, W.; Kuo, M.T.; Feun, L.G. Targeting argininosuccinate synthetase negative melanomas using combination of arginine degrading enzyme and cisplatin. Oncotarget 2015, 6, 6295–6309. [Google Scholar] [CrossRef] [Green Version]
- Wangpaichitr, M.; Wu, C.; Bigford, G.; Theodoropoulos, G.; You, M.; Li, Y.Y.; Verona-Santos, J.; Feun, L.G.; Nguyen, D.M.; Savaraj, N. Combination of arginine deprivation with TRAIL treatment as a targeted-therapy for mesothelioma. Anticancer Res. 2014, 34, 6991–6999. [Google Scholar]
- Locke, M.; Ghazaly, E.; Freitas, M.O.; Mitsinga, M.; Lattanzio, L.; Lo Nigro, C.; Nagano, A.; Wang, J.; Chelala, C.; Szlosarek, P.; et al. Inhibition of the Polyamine Synthesis Pathway Is Synthetically Lethal with Loss of Argininosuccinate Synthase 1. Cell Rep. 2016, 16, 1604–1613. [Google Scholar] [CrossRef] [Green Version]
- Barnett, S.E.; Kenyani, J.; Tripari, M.; Butt, Z.; Grosman, R.; Querques, F.; Shaw, L.; Silva, L.C.; Goate, Z.; Marciniak, S.J.; et al. BAP1 loss is associated with higher ASS1 expression in epithelioid mesothelioma: Implications for therapeutic stratification. Mol. Cancer Res. 2023, 21, 411–427. [Google Scholar] [CrossRef]
- Chalishazar, M.D.; Wait, S.J.; Huang, F.; Ireland, A.S.; Mukhopadhyay, A.; Lee, Y.; Schuman, S.S.; Guthrie, M.R.; Berrett, K.C.; Vahrenkamp, J.M.; et al. MYC-Driven Small-Cell Lung Cancer is Metabolically Distinct and Vulnerable to Arginine Depletion. Clin. Cancer Res. 2019, 25, 5107–5121. [Google Scholar] [CrossRef] [Green Version]
- Zheng, L.; MacKenzie, E.D.; Karim, S.A.; Hedley, A.; Blyth, K.; Kalna, G.; Watson, D.G.; Szlosarek, P.; Frezza, C.; Gottlieb, E. Reversed argininosuccinate lyase activity in fumarate hydratase-deficient cancer cells. Cancer Metab. 2013, 1, 12. [Google Scholar] [CrossRef] [Green Version]
- Noh, E.J.; Kang, S.W.; Shin, Y.J.; Choi, S.H.; Kim, C.G.; Park, I.S.; Wheatley, D.N.; Min, B.H. Arginine deiminase enhances dexamethasone-induced cytotoxicity in human T-lymphoblastic leukemia CCRF-CEM cells. Int. J. Cancer 2004, 112, 502–508. [Google Scholar] [CrossRef] [PubMed]
- Fiedler, T.; Strauss, M.; Hering, S.; Redanz, U.; William, D.; Rosche, Y.; Classen, C.F.; Kreikemeyer, B.; Linnebacher, M.; Maletzki, C. Arginine deprivation by arginine deiminase of Streptococcus pyogenes controls primary glioblastoma growth in vitro and in vivo. Cancer Biol. Ther. 2015, 16, 1047–1055. [Google Scholar] [CrossRef] [Green Version]
- Maletzki, C.; Rosche, Y.; Riess, C.; Scholz, A.; William, D.; Classen, C.F.; Kreikemeyer, B.; Linnebacher, M.; Fiedler, T. Deciphering molecular mechanisms of arginine deiminase-based therapy—Comparative response analysis in paired human primary and recurrent glioblastomas. Chem. Biol. Interact. 2017, 278, 179–188. [Google Scholar] [CrossRef]
- Schwarz, R.; Zitzow, E.; Fiebig, A.; Hering, S.; Humboldt, Y.; Schoenwaelder, N.; Kampfer, N.; Volkmar, K.; Hinz, B.; Kreikemeyer, B.; et al. PEGylation increases antitumoral activity of arginine deiminase of Streptococcus pyogenes. Appl. Microbiol. Biotechnol. 2022, 106, 261–271. [Google Scholar] [CrossRef]
- Riess, C.; Del Moral, K.; Fiebig, A.; Kaps, P.; Linke, C.; Hinz, B.; Rupprecht, A.; Frank, M.; Fiedler, T.; Koczan, D.; et al. Implementation of a combined CDK inhibition and arginine-deprivation approach to target arginine-auxotrophic glioblastoma multiforme cells. Cell Death Dis. 2022, 13, 555. [Google Scholar] [CrossRef]
- Linke, C.; Freitag, T.; Riess, C.; Scheffler, J.V.; Del Moral, K.; Schoenwaelder, N.; Fiedler, T.; Fiebig, A.; Kaps, P.; Dubinski, D.; et al. The addition of arginine deiminase potentiates Mithramycin A-induced cell death in patient-derived glioblastoma cells via ATF4 and cytochrome C. Cancer Cell Int. 2023, 23, 38. [Google Scholar] [CrossRef]
- Chow, J.P.H.; Cai, Y.; Chow, D.T.L.; Chung, S.H.K.; Chau, K.C.; Ng, K.Y.; Leung, O.M.; Wong, R.M.H.; Law, A.W.L.; Leung, Y.O.; et al. A modified arginine-depleting enzyme NEI-01 inhibits growth of pancreatic cancer cells. PLoS ONE 2020, 15, e0231633. [Google Scholar] [CrossRef]
- Cai, Y.; Chow, J.P.H.; Leung, Y.O.; Lu, X.; Yuen, C.H.; Lee, W.L.; Chau, K.C.; Yang, L.L.; Wong, R.M.H.; Lam, J.Y.T.; et al. NEI-01-Induced Arginine Deprivation Has Potent Activity Against Acute Myeloid Leukemia Cells Both In Vitro and In Vivo. Mol. Cancer Ther. 2021, 20, 2218–2227. [Google Scholar] [CrossRef]
- Hall, P.E.; Lewis, R.; Syed, N.; Shaffer, R.; Evanson, J.; Ellis, S.; Williams, M.; Feng, X.; Johnston, A.; Thomson, J.A.; et al. A Phase I Study of Pegylated Arginine Deiminase (Pegargiminase), Cisplatin, and Pemetrexed in Argininosuccinate Synthetase 1-Deficient Recurrent High-grade Glioma. Clin. Cancer Res. 2019, 25, 2708–2716. [Google Scholar] [CrossRef] [Green Version]
- Tomlinson, B.K.; Thomson, J.A.; Bomalaski, J.S.; Diaz, M.; Akande, T.; Mahaffey, N.; Li, T.; Dutia, M.P.; Kelly, K.; Gong, I.Y.; et al. Phase I Trial of Arginine Deprivation Therapy with ADI-PEG 20 Plus Docetaxel in Patients with Advanced Malignant Solid Tumors. Clin. Cancer Res. 2015, 21, 2480–2486. [Google Scholar] [CrossRef] [Green Version]
- Beddowes, E.; Spicer, J.; Chan, P.Y.; Khadeir, R.; Corbacho, J.G.; Repana, D.; Steele, J.P.; Schmid, P.; Szyszko, T.; Cook, G.; et al. Phase 1 Dose-Escalation Study of Pegylated Arginine Deiminase, Cisplatin, and Pemetrexed in Patients With Argininosuccinate Synthetase 1-Deficient Thoracic Cancers. J. Clin. Oncol. 2017, 35, 1778–1785. [Google Scholar] [CrossRef] [Green Version]
- Hall, P.E.; Ready, N.; Johnston, A.; Bomalaski, J.S.; Venhaus, R.R.; Sheaff, M.; Krug, L.; Szlosarek, P.W. Phase II Study of Arginine Deprivation Therapy With Pegargiminase in Patients With Relapsed Sensitive or Refractory Small-cell Lung Cancer. Clin. Lung Cancer 2020, 21, 527–533. [Google Scholar] [CrossRef]
- Szlosarek, P.W.; Wimalasingham, A.G.; Phillips, M.M.; Hall, P.E.; Chan, P.Y.; Conibear, J.; Lim, L.; Rashid, S.; Steele, J.; Wells, P.; et al. Phase 1, pharmacogenomic, dose-expansion study of pegargiminase plus pemetrexed and cisplatin in patients with ASS1-deficient non-squamous non-small cell lung cancer. Cancer Med. 2021, 10, 6642–6652. [Google Scholar] [CrossRef]
- Curley, S.A.; Bomalaski, J.S.; Ensor, C.M.; Holtsberg, F.W.; Clark, M.A. Regression of hepatocellular cancer in a patient treated with arginine deiminase. Hepatogastroenterology 2003, 50, 1214–1216. [Google Scholar]
- Izzo, F.; Marra, P.; Beneduce, G.; Castello, G.; Vallone, P.; De Rosa, V.; Cremona, F.; Ensor, C.M.; Holtsberg, F.W.; Bomalaski, J.S.; et al. Pegylated arginine deiminase treatment of patients with unresectable hepatocellular carcinoma: Results from phase I/II studies. J. Clin. Oncol. 2004, 22, 1815–1822. [Google Scholar] [CrossRef]
- Izzo, F.; Montella, M.; Orlando, A.P.; Nasti, G.; Beneduce, G.; Castello, G.; Cremona, F.; Ensor, C.M.; Holtzberg, F.W.; Bomalaski, J.S.; et al. Pegylated arginine deiminase lowers hepatitis C viral titers and inhibits nitric oxide synthesis. J. Gastroenterol. Hepatol. 2007, 22, 86–91. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.S.; Lu, S.N.; Chao, Y.; Sheen, I.S.; Lin, C.C.; Wang, T.E.; Chen, S.C.; Wang, J.H.; Liao, L.Y.; Thomson, J.A.; et al. A randomised phase II study of pegylated arginine deiminase (ADI-PEG 20) in Asian advanced hepatocellular carcinoma patients. Br. J. Cancer 2010, 103, 954–960. [Google Scholar] [CrossRef] [PubMed]
- Glazer, E.S.; Piccirillo, M.; Albino, V.; Di Giacomo, R.; Palaia, R.; Mastro, A.A.; Beneduce, G.; Castello, G.; De Rosa, V.; Petrillo, A.; et al. Phase II study of pegylated arginine deiminase for nonresectable and metastatic hepatocellular carcinoma. J. Clin. Oncol. 2010, 28, 2220–2226. [Google Scholar] [CrossRef] [PubMed]
- Harding, J.J.; Do, R.K.; Dika, I.E.; Hollywood, E.; Uhlitskykh, K.; Valentino, E.; Wan, P.; Hamilton, C.; Feng, X.; Johnston, A.; et al. A phase 1 study of ADI-PEG 20 and modified FOLFOX6 in patients with advanced hepatocellular carcinoma and other gastrointestinal malignancies. Cancer Chemother. Pharmacol. 2018, 82, 429–440. [Google Scholar] [CrossRef]
- Abou-Alfa, G.K.; Qin, S.; Ryoo, B.Y.; Lu, S.N.; Yen, C.J.; Feng, Y.H.; Lim, H.Y.; Izzo, F.; Colombo, M.; Sarker, D.; et al. Phase III randomized study of second line ADI-PEG 20 plus best supportive care versus placebo plus best supportive care in patients with advanced hepatocellular carcinoma. Ann. Oncol. 2018, 29, 1402–1408. [Google Scholar] [CrossRef]
- Harding, J.J.; Yang, T.S.; Chen, Y.Y.; Feng, Y.H.; Yen, C.J.; Ho, C.L.; Huang, W.T.; El Dika, I.; Akce, M.; Tan, B.; et al. Assessment of pegylated arginine deiminase and modified FOLFOX6 in patients with advanced hepatocellular carcinoma: Results of an international, single-arm, phase 2 study. Cancer 2021, 127, 4585–4593. [Google Scholar] [CrossRef]
- Ascierto, P.A.; Scala, S.; Castello, G.; Daponte, A.; Simeone, E.; Ottaiano, A.; Beneduce, G.; De Rosa, V.; Izzo, F.; Melucci, M.T.; et al. Pegylated arginine deiminase treatment of patients with metastatic melanoma: Results from phase I and II studies. J. Clin. Oncol. 2005, 23, 7660–7668. [Google Scholar] [CrossRef]
- Feun, L.G.; Marini, A.; Walker, G.; Elgart, G.; Moffat, F.; Rodgers, S.E.; Wu, C.J.; You, M.; Wangpaichitr, M.; Kuo, M.T.; et al. Negative argininosuccinate synthetase expression in melanoma tumours may predict clinical benefit from arginine-depleting therapy with pegylated arginine deiminase. Br. J. Cancer 2012, 106, 1481–1485. [Google Scholar] [CrossRef] [PubMed]
- Ott, P.A.; Carvajal, R.D.; Pandit-Taskar, N.; Jungbluth, A.A.; Hoffman, E.W.; Wu, B.W.; Bomalaski, J.S.; Venhaus, R.; Pan, L.; Old, L.J.; et al. Phase I/II study of pegylated arginine deiminase (ADI-PEG 20) in patients with advanced melanoma. Investig. New Drugs 2013, 31, 425–434. [Google Scholar] [CrossRef] [Green Version]
- Yao, S.; Janku, F.; Subbiah, V.; Stewart, J.; Patel, S.P.; Kaseb, A.; Westin, S.N.; Naing, A.; Tsimberidou, A.M.; Hong, D.; et al. Phase 1 trial of ADI-PEG20 plus cisplatin in patients with pretreated metastatic melanoma or other advanced solid malignancies. Br. J. Cancer 2021, 124, 1533–1539. [Google Scholar] [CrossRef]
- Kraehenbuehl, L.; Holland, A.; Armstrong, E.; O’Shea, S.; Mangarin, L.; Chekalil, S.; Johnston, A.; Bomalaski, J.S.; Erinjeri, J.P.; Barker, C.A.; et al. Pilot Trial of Arginine Deprivation Plus Nivolumab and Ipilimumab in Patients with Metastatic Uveal Melanoma. Cancers 2022, 14, 2638. [Google Scholar] [CrossRef]
- Chan, P.Y.; Phillips, M.M.; Ellis, S.; Johnston, A.; Feng, X.; Arora, A.; Hay, G.; Cohen, V.M.L.; Sagoo, M.S.; Bomalaski, J.S.; et al. A Phase 1 study of ADI-PEG20 (pegargiminase) combined with cisplatin and pemetrexed in ASS1-negative metastatic uveal melanoma. Pigment Cell Melanoma Res. 2022, 35, 461–470. [Google Scholar] [CrossRef]
- Szlosarek, P.W.; Steele, J.P.; Nolan, L.; Gilligan, D.; Taylor, P.; Spicer, J.; Lind, M.; Mitra, S.; Shamash, J.; Phillips, M.M.; et al. Arginine Deprivation With Pegylated Arginine Deiminase in Patients With Argininosuccinate Synthetase 1-Deficient Malignant Pleural Mesothelioma: A Randomized Clinical Trial. JAMA Oncol. 2017, 3, 58–66. [Google Scholar] [CrossRef]
- Szlosarek, P.W.; Phillips, M.M.; Pavlyk, I.; Steele, J.; Shamash, J.; Spicer, J.; Kumar, S.; Pacey, S.; Feng, X.; Johnston, A.; et al. Expansion Phase 1 Study of Pegargiminase Plus Pemetrexed and Cisplatin in Patients With Argininosuccinate Synthetase 1-Deficient Mesothelioma: Safety, Efficacy, and Resistance Mechanisms. JTO Clin. Res. Rep. 2020, 1, 100093. [Google Scholar] [CrossRef] [PubMed]
- Tsai, H.J.; Jiang, S.S.; Hung, W.C.; Borthakur, G.; Lin, S.F.; Pemmaraju, N.; Jabbour, E.; Bomalaski, J.S.; Chen, Y.P.; Hsiao, H.H.; et al. A Phase II Study of Arginine Deiminase (ADI-PEG20) in Relapsed/Refractory or Poor-Risk Acute Myeloid Leukemia Patients. Sci. Rep. 2017, 7, 11253. [Google Scholar] [CrossRef] [Green Version]
- Tsai, H.J.; Hsiao, H.H.; Hsu, Y.T.; Liu, Y.C.; Kao, H.W.; Liu, T.C.; Cho, S.F.; Feng, X.; Johnston, A.; Bomalaski, J.S.; et al. Phase I study of ADI-PEG20 plus low-dose cytarabine for the treatment of acute myeloid leukemia. Cancer Med. 2021, 10, 2946–2955. [Google Scholar] [CrossRef]
- Lowery, M.A.; Yu, K.H.; Kelsen, D.P.; Harding, J.J.; Bomalaski, J.S.; Glassman, D.C.; Covington, C.M.; Brenner, R.; Hollywood, E.; Barba, A.; et al. A phase 1/1B trial of ADI-PEG 20 plus nab-paclitaxel and gemcitabine in patients with advanced pancreatic adenocarcinoma. Cancer 2017, 123, 4556–4565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, S.; Janku, F.; Koenig, K.; Tsimberidou, A.M.; Piha-Paul, S.A.; Shi, N.; Stewart, J.; Johnston, A.; Bomalaski, J.; Meric-Bernstam, F.; et al. Phase 1 trial of ADI-PEG 20 and liposomal doxorubicin in patients with metastatic solid tumors. Cancer Med. 2022, 11, 340–347. [Google Scholar] [CrossRef]
- Chang, K.Y.; Chiang, N.J.; Wu, S.Y.; Yen, C.J.; Chen, S.H.; Yeh, Y.M.; Li, C.F.; Feng, X.; Wu, K.; Johnston, A.; et al. Phase 1b study of pegylated arginine deiminase (ADI-PEG 20) plus Pembrolizumab in advanced solid cancers. Oncoimmunology 2021, 10, 1943253. [Google Scholar] [CrossRef]
- Szlosarek, P.W.; Baas, P.; Ceresoli, G.L.; Fennell, D.A.; Gilligan, D.; Johnston, A.; Lee, P.; Mansfield, A.S.; Nolan, L.; Nowak, A.K.; et al. ATOMIC-Meso: A randomized phase 2/3 trial of ADI-PEG20 or placebo with pemetrexed and cisplatin in patients with argininosuccinate synthetase 1-deficient non-epithelioid mesothelioma. J. Clin. Oncol. 2017, 35, TPS8582. [Google Scholar] [CrossRef]
- Tine, B.A.V.; Hirbe, A.C.; Luo, J.; Oppelt, P.J.; Weiss, M.C.; Eulo, V.A.; Toeniskoetter, J.; Haarberg, S.; Abaricia, S.; Ruff, T.; et al. Phase II trial of pegylated arginine deiminase in combination with gemcitabine and docetaxel for the treatment of soft tissue sarcoma. J. Clin. Oncol. 2021, 39, 11508. [Google Scholar] [CrossRef]
- Zou, S.; Wang, X.; Liu, P.; Ke, C.; Xu, S. Arginine metabolism and deprivation in cancer therapy. Biomed. Pharmacother. 2019, 118, 109210. [Google Scholar] [CrossRef]
- Tsui, S.M.; Lam, W.M.; Lam, T.L.; Chong, H.C.; So, P.K.; Kwok, S.Y.; Arnold, S.; Cheng, P.N.; Wheatley, D.N.; Lo, W.H.; et al. Pegylated derivatives of recombinant human arginase (rhArg1) for sustained in vivo activity in cancer therapy: Preparation, characterization and analysis of their pharmacodynamics in vivo and in vitro and action upon hepatocellular carcinoma cell (HCC). Cancer Cell Int. 2009, 9, 9. [Google Scholar] [CrossRef] [Green Version]
- Khoury, O.; Ghazale, N.; Stone, E.; El-Sibai, M.; Frankel, A.E.; Abi-Habib, R.J. Human recombinant arginase I (Co)-PEG5000 [HuArgI (Co)-PEG5000]-induced arginine depletion is selectively cytotoxic to human glioblastoma cells. J. Neuro-Oncol. 2015, 122, 75–85. [Google Scholar] [CrossRef]
- Hinrichs, C.N.; Ingargiola, M.; Käubler, T.; Löck, S.; Temme, A.; Köhn-Luque, A.; Deutsch, A.; Vovk, O.; Stasyk, O.; Kunz-Schughart, L.A. Arginine Deprivation Therapy: Putative Strategy to Eradicate Glioblastoma Cells by Radiosensitization. Mol. Cancer Ther. 2018, 17, 393–406. [Google Scholar] [CrossRef] [Green Version]
- Fultang, L.; Gamble, L.D.; Gneo, L.; Berry, A.M.; Egan, S.A.; De Bie, F.; Yogev, O.; Eden, G.L.; Booth, S.; Brownhill, S.; et al. Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma. Cancer Res. 2019, 79, 611–624. [Google Scholar] [CrossRef] [Green Version]
- Shen, W.; Zhang, X.; Fu, X.; Fan, J.; Luan, J.; Cao, Z.; Yang, P.; Xu, Z.; Ju, D. A novel and promising therapeutic approach for NSCLC: Recombinant human arginase alone or combined with autophagy inhibitor. Cell Death Dis. 2017, 8, e2720. [Google Scholar] [CrossRef] [Green Version]
- Xu, S.; Lam, S.K.; Cheng, P.N.; Ho, J.C. Recombinant human arginase induces apoptosis through oxidative stress and cell cycle arrest in small cell lung cancer. Cancer Sci. 2018, 109, 3471–3482. [Google Scholar] [CrossRef] [Green Version]
- Lam, S.K.; Pong, K.U.; Li, Y.Y.; Xu, S.; Cheng, P.N.; Ho, J.C. Inhibition of ornithine decarboxylase 1 facilitates pegylated arginase treatment in lung adenocarcinoma xenograft models. Oncol. Rep. 2018, 40, 1994–2004. [Google Scholar] [CrossRef] [Green Version]
- Lam, S.K.; Yan, S.; Xu, S.; Pong, K.U.; Cheng, P.N.; Ho, J.C. Endogenous arginase 2 as a potential biomarker for PEGylated arginase 1 treatment in xenograft models of squamous cell lung carcinoma. Oncogenesis 2019, 8, 18. [Google Scholar] [CrossRef]
- Xu, S.; Lam, S.K.; Cheng, P.N.; Ho, J.C. Contactin 1 modulates pegylated arginase resistance in small cell lung cancer through induction of epithelial-mesenchymal transition. Sci. Rep. 2019, 9, 12030. [Google Scholar] [CrossRef] [Green Version]
- Agnello, G.; Alters, S.E.; Rowlinson, S.W. Preclinical safety and antitumor activity of the arginine-degrading therapeutic enzyme pegzilarginase, a PEGylated, cobalt-substituted recombinant human arginase 1. Transl. Res. 2020, 217, 11–22. [Google Scholar] [CrossRef]
- Cheng, P.N.; Lam, T.L.; Lam, W.M.; Tsui, S.M.; Cheng, A.W.; Lo, W.H.; Leung, Y.C. Pegylated recombinant human arginase (rhArg-peg5000mw) inhibits the in vitro and in vivo proliferation of human hepatocellular carcinoma through arginine depletion. Cancer Res. 2007, 67, 309–317. [Google Scholar] [CrossRef] [Green Version]
- Lam, T.L.; Wong, G.K.; Chong, H.C.; Cheng, P.N.; Choi, S.C.; Chow, T.L.; Kwok, S.Y.; Poon, R.T.; Wheatley, D.N.; Lo, W.H.; et al. Recombinant human arginase inhibits proliferation of human hepatocellular carcinoma by inducing cell cycle arrest. Cancer Lett. 2009, 277, 91–100. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Zeng, X.; Wang, S.; Fan, J.; Wang, Z.; Song, P.; Mei, X.; Ju, D. Blocking autophagy enhanced leukemia cell death induced by recombinant human arginase. Tumor Biol. 2016, 37, 6627–6635. [Google Scholar] [CrossRef]
- Kwong-Lam, F.; Chi-Fung, C.G. Vincristine could partly suppress stromal support to T-ALL blasts during pegylated arginase I treatment. Exp. Hematol. Oncol. 2013, 2, 11. [Google Scholar] [CrossRef] [Green Version]
- Mussai, F.; Egan, S.; Higginbotham-Jones, J.; Perry, T.; Beggs, A.; Odintsova, E.; Loke, J.; Pratt, G.; Pong, K.U.; Lo, A.; et al. Arginine dependence of acute myeloid leukemia blast proliferation: A novel therapeutic target. Blood 2015, 125, 2386–2396. [Google Scholar] [CrossRef]
- De Santo, C.; Booth, S.; Vardon, A.; Cousins, A.; Tubb, V.; Perry, T.; Noyvert, B.; Beggs, A.; Ng, M.; Halsey, C.; et al. The arginine metabolome in acute lymphoblastic leukemia can be targeted by the pegylated-recombinant arginase I BCT-100. Int. J. Cancer 2018, 142, 1490–1502. [Google Scholar] [CrossRef] [Green Version]
- Tanios, R.; Bekdash, A.; Kassab, E.; Stone, E.; Georgiou, G.; Frankel, A.E.; Abi-Habib, R.J. 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, 1565–1571. [Google Scholar] [CrossRef]
- Vovk, O.I.; Chen, O.I.; Igumentseva, N.I.; Senchuk, O.Y.; Barska, M.L.; Sybirna, N.O.; Stasyk, O.V. Effects of the combined arginase and canavanine treatment on leukemic cells in vitro and in vivo. Ukr. Biochem. J. 2016, 88, 45–55. [Google Scholar] [CrossRef] [Green Version]
- Lam, T.L.; Wong, G.K.; Chow, H.Y.; Chong, H.C.; Chow, T.L.; Kwok, S.Y.; Cheng, P.N.; Wheatley, D.N.; Lo, W.H.; Leung, Y.C. Recombinant human arginase inhibits the in vitro and in vivo proliferation of human melanoma by inducing cell cycle arrest and apoptosis. Pigment. Cell Melanoma Res. 2011, 24, 366–376. [Google Scholar] [CrossRef]
- Wang, Z.; Shi, X.; Li, Y.; Zeng, X.; Fan, J.; Sun, Y.; Xian, Z.; Zhang, G.; Wang, S.; Hu, H.; et al. Involvement of autophagy in recombinant human arginase-induced cell apoptosis and growth inhibition of malignant melanoma cells. Appl. Microbiol. Biotechnol. 2014, 98, 2485–2494. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Zhao, Z.; Zhang, P.; Li, W.; Wang, D.; Ke, C.; Liu, Y.; Ho, J.C.; Cheng, P.N.; Xu, S. Pegylated Recombinant Human Arginase 1 Induces Autophagy and Apoptosis via the ROS-Activated AKT/mTOR Pathway in Bladder Cancer Cells. Oxidative Med. Cell Longev. 2021, 2021, 5510663. [Google Scholar] [CrossRef]
- Hsueh, E.C.; Knebel, S.M.; Lo, W.H.; Leung, Y.C.; Cheng, P.N.; Hsueh, C.T. Deprivation of arginine by recombinant human arginase in prostate cancer cells. J. Hematol. Oncol. 2012, 5, 17. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Shi, X.; Li, Y.; Fan, J.; Zeng, X.; Xian, Z.; Wang, Z.; Sun, Y.; Wang, S.; Song, P.; et al. Blocking autophagy enhanced cytotoxicity induced by recombinant human arginase in triple-negative breast cancer cells. Cell Death Dis. 2014, 5, e1563. [Google Scholar] [CrossRef] [Green Version]
- Nasreddine, G.; El-Sibai, M.; Abi-Habib, R.J. Cytotoxicity of [HuArgI (co)-PEG5000]-induced arginine deprivation to ovarian Cancer cells is autophagy dependent. Investig. New Drugs 2020, 38, 10–19. [Google Scholar] [CrossRef]
- El-Mais, N.; Fakhoury, I.; Abdellatef, S.; Abi-Habib, R.; El-Sibai, M. Human recombinant arginase I [HuArgI (Co)-PEG5000]-induced arginine depletion inhibits ovarian cancer cell adhesion and migration through autophagy-mediated inhibition of RhoA. J. Ovarian Res. 2021, 14, 13. [Google Scholar] [CrossRef]
- Lam, S.K.; Li, Y.Y.; Xu, S.; Leung, L.L.; Pong, K.U.; Zheng, Y.F.; Cheng, P.N.; Ho, J.C. Growth suppressive effect of pegylated arginase in malignant pleural mesothelioma xenografts. Respir. Res. 2017, 18, 80. [Google Scholar] [CrossRef] [Green Version]
- Chung, S.-F.; Kim, C.-F.; Tam, S.-Y.; Choi, M.-C.; So, P.-K.; Wong, K.-Y.; Leung, Y.-C.; Lo, W.-H. A bioengineered arginine-depleting enzyme as a long-lasting therapeutic agent against cancer. Appl. Microbiol. Biotechnol. 2020, 104, 3921–3934. [Google Scholar] [CrossRef]
- Yau, T.; Cheng, P.N.; Chan, P.; Chan, W.; Chen, L.; Yuen, J.; Pang, R.; Fan, S.T.; Poon, R.T. A phase 1 dose-escalating study of pegylated recombinant human arginase 1 (Peg-rhArg1) in patients with advanced hepatocellular carcinoma. Investig. New Drugs 2013, 31, 99–107. [Google Scholar] [CrossRef] [Green Version]
- Yau, T.; Cheng, P.N.; Chan, P.; Chen, L.; Yuen, J.; Pang, R.; Fan, S.T.; Wheatley, D.N.; Poon, R.T. Preliminary efficacy, safety, pharmacokinetics, pharmacodynamics and quality of life study of pegylated recombinant human arginase 1 in patients with advanced hepatocellular carcinoma. Investig. New Drugs 2015, 33, 496–504. [Google Scholar] [CrossRef]
- Chan, S.L.; Cheng, P.N.M.; Liu, A.M.; Chan, L.L.; Li, L.; Chu, C.M.; Chong, C.C.N.; Lau, Y.M.; Yeo, W.; Ng, K.K.C.; et al. A phase II clinical study on the efficacy and predictive biomarker of pegylated recombinant arginase on hepatocellular carcinoma. Investig. New Drugs 2021, 39, 1375–1382. [Google Scholar] [CrossRef]
- Yau, T.; Cheng, P.N.M.; Chiu, J.; Kwok, G.G.W.; Leung, R.; Liu, A.M.; Cheung, T.T.; Ng, C.T. A phase 1 study of pegylated recombinant arginase (PEG-BCT-100) in combination with systemic chemotherapy (capecitabine and oxaliplatin)[PACOX] in advanced hepatocellular carcinoma patients. Investig. New Drugs 2022, 40, 314–321. [Google Scholar] [CrossRef]
- Cheng, P.N.M.; Liu, A.M.; Bessudo, A.; Mussai, F. Safety, PK/PD and preliminary anti-tumor activities of pegylated recombinant human arginase 1 (BCT-100) in patients with advanced arginine auxotrophic tumors. Investig. New Drugs 2021, 39, 1633–1640. [Google Scholar] [CrossRef]
- De Santo, C.; Cheng, P.; Beggs, A.; Egan, S.; Bessudo, A.; Mussai, F. Metabolic therapy with PEG-arginase induces a sustained complete remission in immunotherapy-resistant melanoma. J. Hematol. Oncol. 2018, 11, 68. [Google Scholar] [CrossRef]
- Mussai, F.; De Santo, C.; Cheng, P.; Thomas, I.F.; Ariti, C.; Upton, L.; Scarpa, U.; Stavrou, V.; Sydenham, M.; Burnett, A.K.; et al. A randomised evaluation of low-dose Ara-C plus pegylated recombinant arginase BCT-100 versus low dose Ara-C in older unfit patients with acute myeloid leukaemia: Results from the LI-1 trial. Br. J. Haematol. 2023, 200, 573–578. [Google Scholar] [CrossRef]
- Li, J.; Meng, Y.; Wu, X.; Sun, Y. Polyamines and related signaling pathways in cancer. Cancer Cell Int. 2020, 20, 539. [Google Scholar] [CrossRef]
- Wolf, C.; Brüss, M.; Hänisch, B.; Göthert, M.; von Kügelgen, I.; Molderings, G.J. Molecular basis for the antiproliferative effect of agmatine in tumor cells of colonic, hepatic, and neuronal origin. Mol. Pharmacol. 2007, 71, 276–283. [Google Scholar] [CrossRef] [Green Version]
- Philip, R.; Campbell, E.; Wheatley, D.N. Arginine deprivation, growth inhibition and tumour cell death: 2. Enzymatic degradation of arginine in normal and malignant cell cultures. Br. J. Cancer 2003, 88, 613–623. [Google Scholar] [CrossRef]
- Leung, Y.-C.; Wei, X.-L. Arginine decarboxylase inhibits human colorectal cancer cells by inducing cell cycle arrest and apoptosis. Eur. J. Cancer 2012, 48, 25–26. [Google Scholar]
- Verma, A.; Lam, Y.M.; Leung, Y.C.; Hu, X.; Chen, X.; Cheung, E.; Tam, K.Y. Combined use of arginase and dichloroacetate exhibits anti-proliferative effects in triple negative breast cancer cells. J. Pharm. Pharmacol. 2019, 71, 306–315. [Google Scholar] [CrossRef]
- Chung, S.F.; Kim, C.F.; Chow, H.Y.; Chong, H.C.; Tam, S.Y.; Leung, Y.C.; Lo, W.H. Recombinant Bacillus caldovelox Arginase Mutant (BCA-M) Induces Apoptosis, Autophagy, Cell Cycle Arrest and Growth Inhibition in Human Cervical Cancer Cells. Int. J. Mol. Sci. 2020, 21, 7445. [Google Scholar] [CrossRef]
- Chung, S.F.; Kim, C.F.; Kwok, S.Y.; Tam, S.Y.; Chen, Y.W.; Chong, H.C.; Leung, S.L.; So, P.K.; Wong, K.Y.; Leung, Y.C.; et al. Mono-PEGylation of a Thermostable Arginine-Depleting Enzyme for the Treatment of Lung Cancer. Int. J. Mol. Sci. 2020, 21, 4234. [Google Scholar] [CrossRef]
- Chung, S.F.; Tam, S.Y.; Kim, C.F.; Chong, H.C.; Lee, L.M.; Leung, Y.C. Mono-PEGylated thermostable Bacillus caldovelox arginase mutant (BCA-M-PEG20) induces apoptosis, autophagy, cell cycle arrest and growth inhibition in gastric cancer cells. Investig. New Drugs 2022, 40, 895–904. [Google Scholar] [CrossRef]
- Werner, A.; Pieh, D.; Echchannaoui, H.; Rupp, J.; Rajalingam, K.; Theobald, M.; Closs, E.I.; Munder, M. Cationic Amino Acid Transporter-1-Mediated Arginine Uptake Is Essential for Chronic Lymphocytic Leukemia Cell Proliferation and Viability. Front. Oncol. 2019, 9, 1268. [Google Scholar] [CrossRef]
- Bröer, S. Amino Acid Transporters as Targets for Cancer Therapy: Why, Where, When, and How. Int. J. Mol. Sci. 2020, 21, 6156. [Google Scholar] [CrossRef]
- Saito, Y.; Soga, T. Amino acid transporters as emerging therapeutic targets in cancer. Cancer Sci. 2021, 112, 2958–2965. [Google Scholar] [CrossRef]
- Nagasawa, I.; Koido, M.; Tani, Y.; Tsukahara, S.; Kunimasa, K.; Tomida, A. Disrupting ATF4 Expression Mechanisms Provides an Effective Strategy for BRAF-Targeted Melanoma Therapy. iScience 2020, 23, 101028. [Google Scholar] [CrossRef]
- Chantranupong, L.; Scaria, S.M.; Saxton, R.A.; Gygi, M.P.; Shen, K.; Wyant, G.A.; Wang, T.; Harper, J.W.; Gygi, S.P.; Sabatini, D.M. The CASTOR Proteins Are Arginine Sensors for the mTORC1 Pathway. Cell 2016, 165, 153–164. [Google Scholar] [CrossRef] [Green Version]
- Buel, G.R.; Dang, H.Q.; Asara, J.M.; Blenis, J.; Mutvei, A.P. Prolonged deprivation of arginine or leucine induces PI3K/Akt-dependent reactivation of mTORC1. J. Biol. Chem. 2022, 298, 102030. [Google Scholar] [CrossRef]
- Li, T.; Wang, X.; Ju, E.; da Silva, S.R.; Chen, L.; Zhang, X.; Wei, S.; Gao, S.J. RNF167 activates mTORC1 and promotes tumorigenesis by targeting CASTOR1 for ubiquitination and degradation. Nat. Commun. 2021, 12, 1055. [Google Scholar] [CrossRef]
- Rosenthal, G.A. The biological effects and mode of action of L-canavanine, a structural analogue of L-arginine. Q. Rev. Biol. 1977, 52, 155–178. [Google Scholar] [CrossRef]
- Bobak, Y.; Kurlishchuk, Y.; Vynnytska-Myronovska, B.; Grydzuk, O.; Shuvayeva, G.; Redowicz, M.J.; Kunz-Schughart, L.A.; Stasyk, O. Arginine deprivation induces endoplasmic reticulum stress in human solid cancer cells. Int. J. Biochem. Cell Biol. 2016, 70, 29–38. [Google Scholar] [CrossRef]
- Chen, O.; Manig, F.; Lehmann, L.; Sorour, N.; Löck, S.; Yu, Z.; Dubrovska, A.; Baumann, M.; Kessler, B.M.; Stasyk, O.; et al. Dual role of ER stress in response to metabolic co-targeting and radiosensitivity in head and neck cancer cells. Cell. Mol. Life Sci. 2021, 78, 3021–3044. [Google Scholar] [CrossRef]
- Field, G.C.; Pavlyk, I.; Szlosarek, P.W. Bench-to-Bedside Studies of Arginine Deprivation in Cancer. Molecules 2023, 28, 2150. [Google Scholar] [CrossRef]
- Fultang, L.; Vardon, A.; De Santo, C.; Mussai, F. Molecular basis and current strategies of therapeutic arginine depletion for cancer. Int. J. Cancer 2016, 139, 501–509. [Google Scholar] [CrossRef] [Green Version]
- Semenza, G.L. Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharmacol. Sci. 2012, 33, 207–214. [Google Scholar] [CrossRef] [Green Version]
- Bouleftour, W.; Rowinski, E.; Louati, S.; Sotton, S.; Wozny, A.S.; Moreno-Acosta, P.; Mery, B.; Rodriguez-Lafrasse, C.; Magne, N. A Review of the Role of Hypoxia in Radioresistance in Cancer Therapy. Med. Sci. Monit. 2021, 27, e934116. [Google Scholar] [CrossRef]
- Burrows, N.; Cane, G.; Robson, M.; Gaude, E.; Howat, W.J.; Szlosarek, P.W.; Pedley, R.B.; Frezza, C.; Ashcroft, M.; Maxwell, P.H. Hypoxia-induced nitric oxide production and tumour perfusion is inhibited by pegylated arginine deiminase (ADI-PEG20). Sci. Rep. 2016, 6, 22950. [Google Scholar] [CrossRef] [Green Version]
- Taouis, K.; Driouch, K.; Lidereau, R.; Lallemand, F. Molecular Functions of WWOX Potentially Involved in Cancer Development. Cells 2021, 10, 1051. [Google Scholar] [CrossRef]
- Abu-Remaileh, M.; Aqeilan, R.I. Tumor suppressor WWOX regulates glucose metabolism via HIF1α modulation. Cell Death Differ. 2014, 21, 1805–1814. [Google Scholar] [CrossRef] [Green Version]
- Bendinelli, P.; Maroni, P.; Matteucci, E.; Luzzati, A.; Perrucchini, G.; Desiderio, M.A. Hypoxia inducible factor-1 is activated by transcriptional co-activator with PDZ-binding motif (TAZ) versus WWdomain-containing oxidoreductase (WWOX) in hypoxic microenvironment of bone metastasis from breast cancer. Eur. J. Cancer 2013, 49, 2608–2618. [Google Scholar] [CrossRef]
- Abu-Remaileh, M.; Khalaileh, A.; Pikarsky, E.; Aqeilan, R.I. WWOX controls hepatic HIF1α to suppress hepatocyte proliferation and neoplasia. Cell Death Dis. 2018, 9, 511. [Google Scholar] [CrossRef] [Green Version]
- Maroni, P.; Matteucci, E.; Bendinelli, P.; Desiderio, M.A. Functions and Epigenetic Regulation of Wwox in Bone Metastasis from Breast Carcinoma: Comparison with Primary Tumors. Int. J. Mol. Sci. 2017, 18, 75. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.Y.; Wu, C.; Chen, S.M.; Shah, S.S.; Wangpaichitr, M.; Feun, L.G.; Kuo, M.T.; Suarez, M.; Prince, J.; Savaraj, N. BRAF inhibitor resistance enhances vulnerability to arginine deprivation in melanoma. Oncotarget 2016, 7, 17665–17680. [Google Scholar] [CrossRef]
- Li, Y.Y.; Wu, C.; Shah, S.S.; Chen, S.M.; Wangpaichitr, M.; Kuo, M.T.; Feun, L.G.; Han, X.; Suarez, M.; Prince, J.; et al. Degradation of AMPK-α1 sensitizes BRAF inhibitor-resistant melanoma cells to arginine deprivation. Mol. Oncol. 2017, 11, 1806–1825. [Google Scholar] [CrossRef] [Green Version]
Cancer Type | ASS1 Expression | Method | Reference |
---|---|---|---|
Glioma | Reduced in tumor | IHC and WB | [23,24] |
Lung cancer | Reduced in SCLC and LCNEC | IHC | [25,26] |
Head and neck cancer | Reduced in tumor | IHC | [27] |
Hepatocellular carcinoma | Reduced in tumor | cDNA dot blotting, IHC and WB | [21,28] |
Pancreatic cancer | Reduced in tumor | IHC and cDNA dot blotting | [21,29] |
Renal cancer | Reduced in tumor | IHC and cDNA dot blotting | [21,30] |
Bladder cancer | Reduced in tumor | IHC and RT-qPCR | [31,32] |
Prostate cancer | Reduced in tumor | cDNA dot blotting | [21] |
Breast cancer | Reduced in tumor | RT-qPCR | [33] |
Sarcoma | Reduced in tumor | cDNA microarray and IHC | [34] |
Ovarian cancer | Reduced in tumor | cDNA dot blotting and IHC | [21,35] |
Melanoma | Reduced in tumor | cDNA dot blotting | [21] |
Lymphoma and leukemia | Reduced in tumor | IHC, WB and RT-qPCR | [36,37] |
Mesothelioma | Reduced in tumor | cDNA dot blotting | [21] |
Benefit | Disadvantage and Challenge |
---|---|
Lower toxicity | Only effective in individuals with specific biomarkers (e.g., ASS1-negative) |
Fewer side effects | |
Good tolerability | |
Potential add-on therapy |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chu, Y.-D.; Lai, M.-W.; Yeh, C.-T. Unlocking the Potential of Arginine Deprivation Therapy: Recent Breakthroughs and Promising Future for Cancer Treatment. Int. J. Mol. Sci. 2023, 24, 10668. https://doi.org/10.3390/ijms241310668
Chu Y-D, Lai M-W, Yeh C-T. Unlocking the Potential of Arginine Deprivation Therapy: Recent Breakthroughs and Promising Future for Cancer Treatment. International Journal of Molecular Sciences. 2023; 24(13):10668. https://doi.org/10.3390/ijms241310668
Chicago/Turabian StyleChu, Yu-De, Ming-Wei Lai, and Chau-Ting Yeh. 2023. "Unlocking the Potential of Arginine Deprivation Therapy: Recent Breakthroughs and Promising Future for Cancer Treatment" International Journal of Molecular Sciences 24, no. 13: 10668. https://doi.org/10.3390/ijms241310668