Open Access

Recombinant Methioninase Lowers the Effective Dose of Regorafenib Against Colon-Cancer Cells: A Strategy for Widespread Clinical Use of a Toxic Drug

CHOOBIN BRAD BARSAM 1
KUBOTA YUTARO 1 2 3
HAN QINGHONG 1
ARDJMAND DANIEL 1
MORINAGA SEI 1 2
MIZUTA KOHEI 1 2
BOUVET MICHAEL 2
TSUNODA TAKUYA 3
  &  
HOFFMAN ROBERT M. 1 2

1AntiCancer Inc., San Diego, CA, U.S.A.

2Department of Surgery, University of California, San Diego, CA, U.S.A.

3Division of Internal Medicine, Department of Medical Oncology, Showa University School of Medicine, Tokyo, Japan

Cancer Diagnosis & Prognosis Nov-Dec; 3(6): 655-659 DOI: 10.21873/cdp.10268
Received 24 August 2023 | Revised 03 December 2024 | Accepted 26 September 2023
Corresponding author
Robert M. Hoffman, Ph.D., AntiCancer Inc, 7917 Ostrow St, San Diego, CA, 92111, U.S.A. Tel: +1 6198852284, email: all@anticancer.com

Abstract

Background/Aim: Regorafenib is a multi-kinase inhibitor, targeting vascular endothelial growth factor receptor 2, fibroblast growth factor receptor 1 and other oncogenic kinases. Regorafenib has efficacy in metastatic colon cancer, but has severe dose-limiting toxicities which cause patients to stop taking the drug. The aim of the present study was to determine if recombinant methioninase (rMETase) could lower the effective concentration of regorafenib in vitro against a colorectal-cancer cell line. Materials and Methods: Firstly, we examined the half-maximal inhibitory concentration (IC50) of regorafenib alone and rMETase alone for the HCT-116 human colorectal-cancer cell line. After that, using the IC50 concentration of each drug, we investigated the efficacy of the combination of regorafenib and rMETase. Results: While both methioninase alone (IC50=0.61 U/ml) and regorafenib alone (IC50=2.26 U/ml) inhibited the viability of HCT-116 cells, the combination of the two agents was more than twice as effective as either alone. Addition of rMETase at 0.61 U/ml lowered the IC50 of regorafenib from 2.26 μM to 1.46 μM. Conclusion: rMETase and regorafenib are synergistic, giving rise to the possibility of lowering the effective dose of regorafenib in patients, thereby reducing its severe toxicity, allowing more cancer patients to be treated with regorafenib.
Keywords: Regorafenib, multi-kinase inhibitor, toxicity, Methionine addiction, Hoffman effect, recombinant, methioninase, rMETase, combination treatment, IC50, synergy, colon cancer cells, HCT-116

Methionine addiction is a fundamental and general hallmark of cancer, termed the Hoffman effect (1-5). Methionine restriction was shown to increase the efficacy of chemotherapy drugs on cancer cells almost 40 years ago (2,3). Since then, methionine restriction using recombinant methioninase (rMETase), cloned from Pseudomonas putida, has been shown to increase the efficacy of all cytotoxic chemotherapy drugs tested (2). rMETase targets the methionine addiction of cancer (1-5), and selectively arrests cancer cells in late-S/G2 phase at which many cytotoxic chemotherapy drugs are active (6-9).

However, rMETase has not been tested in combination with a multiple kinase inhibitor, such as regorafenib. Regorafenib has increased the overall survival of patients with metastatic colorectal cancer (10) and gastro-intestinal stromal tumor (11) with whom first- or second-line therapies have failed. However, the toxicity of regorafenib appears within 3-4 days of the start of treatment and includes severe skin rash, diarrhea, hypertension and fatigue (12). These severe side-effects make it difficult to start regorafenib at the recommended dose of 160 mg/day. We previously reported that 160 mg/day regorafenib increased blood levels of M2 and M5, which are active metabolites of regorafenib, responsible for the severe side-effects. Thus the dose intensity of regorafenib was then reduced, thereby reducing its efficacy (13). Recently, clinical trials showed that starting at a low dose of regorafenib (80 or 120 mg/day) can reduce side-effects and increase dose intensity (14,15). According to these results, the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) recommends a starting dose of 80 mg/day rather than the usual 160 mg/day dose, with dose escalation depending on side-effects (16). However, low-dose regorafenib can also reduce efficacy for some patients.

In the present study, we tested regorafenib in combination with rMETase on a human colon cancer cell line in vitro to determine whether rMETase can reduce the effective dosage of regorafenib.

Materials and Methods

Cell culture. The HCT-116 human colon cancer cell line was acquired from the American Type Culture Collection (Manassas, VA, USA). The cells were grown in Dulbecco’s minimum essential medium (DMEM) with 10% fetal bovine serum and 100 IU/ml of penicillin/ streptomycin.

rMETase production and formulation. rMETase was produced at AntiCancer Inc. (San Diego, CA, USA) by fermentation of recombinant Escherichia coli transformed with the methioninase gene from Pseudomonas putida. rMETase was purified using a high-yield method, including a 60˚C thermal step, polyethylene-glycol precipitation, and diethylaminoethyl-sepharose fast-flow column chromatography (17).

Cell viability testing. HCT-116 cells were cultivated at subconfluence overnight in Dulbecco’s modified Eagle’s medium in 96-well plates (1.0×103 cells per well). The following day, HCT-116 cells were treated with concentrations of rMETase ranging from 0.125 to 8 U/ml or regorafenib ranging from 0.25 μM to 16 μM. Regorafenib was purchased from MedChemExpress (Monmouth Junction, NJ, USA). After 72 h of treatment, cell viability was assessed using the Cell Counting Kit-8 (Dojindo Laboratory, Kumamoto, Japan) with the WST-8 reagent.

ImageJ version 1.53 (National Institutes of Health, Bethesda, MD, USA) was applied to calculate IC50 values and sensitivity curves. After calculating the half-maximal inhibitory concentration (IC50) for rMETase and regorafenib, the IC50 concentrations of both drugs were used to determine the synergistic efficacy of the combination of the drugs. Finally, we repeated the IC50 study for regorafenib on HCT-116 cells treated in combination with the IC50 concentration of rMETase to examine whether methionine restriction increased the efficacy of regorafenib. Each experiment was carried out in triplicate.

Statistics. GraphPad Prism 9.4.0 (GraphPad Software, Inc., San Diego, CA, USA) was used to conduct all statistical analyses. Tukey’s multiple comparison test was performed for the parametric test of comparison between groups. All data are presented as the mean and standard deviation. The significance level was p≤0.05.

Results

Determination of the IC50 of rMETase alone and regorafenib alone and efficacy of their combination on HCT-116 cells in vitro. We first evaluated the sensitivity to rMETase alone and regorafenib alone of HCT-116 cells in vitro, and IC50 values were calculated. The IC50 of rMETase on HCT-116 cells was 0.61 U/ml and the IC50 of regorafenib was 2.26 μM (Figure 1 and Figure 2). The combination of rMETase and regorafenib at their IC50 highly reduced the viability of HCT-116 cells in comparison to either agent alone at its IC50 (Figure 3).

Reduction of the IC50 of regorafenib in combination with the IC50 of rMETase. The IC50 of regorafenib against HCT-116 was reduced from 2.26 μM to 1.46 μM when cells were treated in the presence of rMETase at its IC50 (Figure 4).

Discussion

Regorafenib is a third-line chemotherapy drug for metastatic colorectal cancer and gastro-intestinal stromal tumor, used after the failure of first- or second-line therapy. The problem with regorafenib is its dose-limiting toxicity. The results of the present study suggest that rMETase, when combined with regorafenib, can reduce the effective dose of regorafenib and thereby also reduce its toxicity. Previously, we showed in an orthotopic mouse model of osteosarcoma that combining rMETase with cisplatinum reduced its effective dose by 50% (18). A similar effect is predicted for the combination of regorafenib and rMETase in future in vivo experiments.

Regorafenib is a multikinase inhibitor of angiogenic, stromal, and oncogenic-receptor tyrosine kinases. In an in vitro study using the human colorectal cancer cell line HT-29, regorafenib increased the intra-cellular S-adenosyl-L-methionine (SAM) level by 34.5% compared to the control (19). SAM is the only provider of methyl groups to DNA, RNA, histones and other proteins. This suggests that regorafenib inhibits transmethylation reactions which use SAM. Methionine depletion reduces SAM levels in cells, further reducing transmethylation reactions which are elevated in cancer cells (4,20-29). Therefore, the combination of rMETase and regorafenib had synergistic efficacy in the present study, giving rise to the possibility of lowering the effective dose of regorafenib in patients, thereby reducing its severe toxicity, and increasing the use of regorafenib in the clinic. rMETase is effective because it targets methionine addiction, the fundamental hallmark of cancer (1-5,20-33).

Conflicts of Interest

The Authors declare no competing interests regarding this work.

Authors’ Contributions

BBC and YK performed experiments. BBC, YK, and RMH wrote the article. QH provided methioninase. DA, SM, KM, MB, and TT critically reviewed the article.

Acknowledgements

This paper is dedicated to the memory of A. R. Moossa, MD, Sun Lee, MD, Gordon H. Sato, PhD, Professor Li Jiaxi, Masaki Kitajima, MD, Shigeo Yagi, PhD, Jack Geller, MD, Joseph R Bertino, MD, and J.A.R. Mead, PhD. The Robert M Hoffman Foundation for Cancer Research provided funds for this study.

References

1 Hoffman RM & Erbe RW High in vivo rates of methionine biosynthesis in transformed human and malignant rat cells auxotrophic for methionine. Proc Natl Acad Sci USA. 73(5) 1523 - 1527 1976. DOI: 10.1073/pnas.73.5.1523
2 Kubota Y Han Q Aoki Y Masaki N Obara K Hamada K Hozumi C Wong ACW Bouvet M Tsunoda T & Hoffman RM Synergy of combining methionine restriction and chemotherapy: The disruptive next generation of cancer treatment. Cancer Diagn Progn. 3(3) 272 - 281 2023. DOI: 10.21873/cdp.10212
3 Stern PH & Hoffman RM Enhanced in vitro selective toxicity of chemotherapeutic agents for human cancer cells based on a metabolic defect. J Natl Cancer Inst. 76(4) 629 - 39 1986. DOI: 10.1093/jnci/76.4.629
4 Wang Z Yip LY Lee JHJ Wu Z Chew HY Chong PKW Teo CC Ang HY Peh KLE Yuan J Ma S Choo LSK Basri N Jiang X Yu Q Hillmer AM Lim WT Lim TKH Takano A Tan EH Tan DSW Ho YS Lim B & Tam WL Methionine is a metabolic dependency of tumor-initiating cells. Nat Med. 25(5) 825 - 837 2019. DOI: 10.1038/s41591-019-0423-5
5 Kaiser P Methionine dependence of cancer. Biomolecules. 10(4) 568 2020. DOI: 10.3390/biom10040568
6 Yano S Li S Han Q Tan Y Bouvet M Fujiwara T & Hoffman RM Selective methioninase-induced trap of cancer cells in S/G2 phase visualized by FUCCI imaging confers chemosensitivity. Oncotarget. 5(18) 8729 - 8736 2014. DOI: 10.18632/oncotarget.2369
7 Yoshioka T Wada T Uchida N Maki H Yoshida H Ide N Kasai H Hojo K Shono K Maekawa R Yagi S Hoffman RM & Sugita K Anticancer efficacy in vivo and in vitro, synergy with 5-fluorouracil, and safety of recombinant methioninase. Cancer Res. 58(12) 2583 - 2587 1998.
8 Tan Y Sun X Xu M Tan X Sasson A Rashidi B Han Q Tan X Wang X An Z Sun FX & Hoffman RM Efficacy of recombinant methioninase in combination with cisplatin on human colon tumors in nude mice. Clin Cancer Res. 5 2157 - 2163 1999.
9 Hoffman RM & Jacobsen SJ Reversible growth arrest in simian virus 40- transformed human fibroblasts. Proc Natl Acad Sci USA. 77(12) 7306 - 7310 1980. PMID: 6261250. DOI: 10.1073/pnas.77.12.7306
10 Grothey A Van Cutsem E Sobrero A Siena S Falcone A Ychou M Humblet Y Bouché O Mineur L Barone C Adenis A Tabernero J Yoshino T Lenz H-J Goldberg RM Sargent DJ Cihon F Cupit L Wagner A Laurent D & CORRECT Study Group Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet (London, England). 381 303 - 312 2013. DOI: 10.1016/S0140-6736(12)61900-X
11 Demetri GD Reichardt P Kang YK Blay JY Rutkowski P Gelderblom H Hohenberger P Leahy M von Mehren M Joensuu H Badalamenti G Blackstein M Le Cesne A Schöffski P Maki RG Bauer S Nguyen BB Xu J Nishida T Chung J Kappeler C Kuss I Laurent D Casali PG & GRID study investigators Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 381(9863) 295 - 302 2013. DOI: 10.1016/S0140-6736(12)61857-1
12 Krishnamoorthy SK Relias V Sebastian S Jayaraman V & Saif MW Management of regorafenib-related toxicities: a review. Therap Adv Gastroenterol. 8(5) 285 - 297 2015. DOI: 10.1177/1756283X15580743
13 Kubota Y Fujita K Takahashi T Sunakawa Y Ishida H Hamada K Ichikawa W Tsunoda T Shimada K Masuo Y Kato Y & Sasaki Y Higher systemic exposure to unbound active metabolites of regorafenib is associated with short progression-free survival in colorectal cancer patients. Clin Pharmacol Ther. 108(3) 586 - 595 2020. DOI: 10.1002/cpt.1810
14 Bekaii-Saab TS Ou FS Ahn DH Boland PM Ciombor KK Heying EN Dockter TJ Jacobs NL Pasche BC Cleary JM Meyers JP Desnoyers RJ McCune JS Pedersen K Barzi A Chiorean EG Sloan J Lacouture ME Lenz HJ & Grothey A Regorafenib dose-optimisation in patients with refractory metastatic colorectal cancer (ReDOS): a randomised, multicentre, open-label, phase 2 study. Lancet Oncol. 20(8) 1070 - 1082 2019. DOI: 10.1016/S1470-2045(19)30272-4
15 Suzuki T Sukawa Y Imamura CK Masuishi T Satake H Kumekawa Y Funakoshi S Kotaka M Horie Y Kawai S Okuda H Terazawa T Kondoh C Kato K Yoshimura K Ishikawa H Hamamoto Y Boku N Takaishi H & Kanai T A Phase II study of regorafenib with a lower starting dose in patients with metastatic colorectal cancer: Exposure–toxicity analysis ofunbound regorafenib and its active metabolites (RESET trial). Clin Colorectal Cancer. 19(1) 13 - 21.e3 2020. DOI: 10.1016/j.clcc.2019.10.004
16 NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines): Colon Cancer Version 3, 2023. National Comprehensive Cancer Network, Plymouth Meeting, PA, USA. Available at:https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf.
17 Tan Y Xu M Tan X Tan X Wang X Saikawa Y Nagahama T Sun X Lenz M & Hoffman RM Overexpression and large-scale production of recombinantl-methionine-α-deamino-γ-mercaptomethane-lyase for novel anticancer therapy. Protein Expr Purif. 9(2) 233 - 245 1997. DOI: 10.1006/prep.1996.0700
18 Masaki N Han Q Wu NF Samonte C Wu J Hozumi C Obara K Kubota Y Aoki Y Miyazaki J & Hoffman RM Oral-recombinant methioninase lowers the effective dose and eliminates toxicity of cisplatinum for primary osteosarcoma of the mammary gland in a patient-derived orthotopic xenograft mouse model. In Vivo. 36(6) 2598 - 2603 2022. DOI: 10.21873/invivo.12994
19 Ogihara S Komatsu T Itoh Y Miyake Y Suzuki T Yanagi K Kimura Y Ueno T Hanaoka K Kojima H Okabe T Nagano T & Urano Y Metabolic-pathway-oriented screening targeting S-Adenosyl-l-methionine reveals the epigenetic remodeling activities of naturally occurring catechols. J Am Chem Soc. 142(1) 21 - 26 2020. DOI: 10.1021/jacs.9b08698
20 Coalson DW Mecham JO Stern PH & Hoffman RM Reduced availability of endogenously synthesized methionine for S-adenosylmethionine formation in methionine-dependent cancer cells. Proc Natl Acad Sci USA. 79(14) 4248 - 4251 1982. DOI: 10.1073/pnas.79.14.4248
21 Stern PH Mecham JO Wallace CD & Hoffman RM Reduced free-methionine in methionine-dependent SV40-transformed human fibroblasts synthesizing apparently normal amounts of methionine. J Cell Physiol. 117(1) 9 - 14 1983. DOI: 10.1002/jcp.1041170103
22 Stern PH & Hoffman RM Elevated overall rates of transmethylation in cell lines from diverse human tumors. In Vitro. 20(8) 663 - 670 1984. DOI: 10.1007/BF02619617
23 Yamamoto J Han Q Inubushi S Sugisawa N Hamada K Nishino H Miyake K Kumamoto T Matsuyama R Bouvet M Endo I & Hoffman RM Histone methylation status of H3K4me3 and H3K9me3 under methionine restriction is unstable in methionine-addicted cancer cells, but stable in normal cells. Biochem Biophys Res Commun. 533(4) 1034 - 1038 2020. DOI: 10.1016/j.bbrc.2020.09.108
24 Aoki Y Han Q Tome Y Yamamoto J Kubota Y Masaki N Obara K Hamada K Wang JD Inubushi S Bouvet M Clarke SG Nishida K & Hoffman RM Reversion of methionine addiction of osteosarcoma cells to methionine independence results in loss of malignancy, modulation of the epithelial-mesenchymal phenotype and alteration of histone-H3 lysine-methylation. Front Oncol. 12 1009548 2022. DOI: 10.3389/fonc.2022.1009548
25 Yamamoto J Inubushi S Han Q Tashiro Y Sugisawa N Hamada K Aoki Y Miyake K Matsuyama R Bouvet M Clarke SG Endo I & Hoffman RM Linkage of methionine addiction, histone lysine hypermethylation, and malignancy. iScience. 25(4) 104162 2022. DOI: 10.1016/j.isci.2022.104162
26 Aoki Y Tome Y Han Q Yamamoto J Hamada K Masaki N Kubota Y Bouvet M Nishida K & Hoffman RM Deletion of MTAP highly sensitizes osteosarcoma cells to methionine restriction with recombinant methioninase. Cancer Genomics Proteomics. 19(3) 299 - 304 2022. DOI: 10.21873/cgp.20321
27 Yamamoto J Aoki Y Inubushi S Han Q Hamada K Tashiro Y Miyake K Matsuyama R Bouvet M Clarke SG Endo I & Hoffman RM Extent and instability of trimethylation of histone H3 lysine increases with degree of malignancy and methionine addiction. Cancer Genomics Proteomics. 19(1) 12 - 18 2022. DOI: 10.21873/cgp.20299
28 Aoki Y Tome Y Han Q Yamamoto J Hamada K Masaki N Bouvet M Nishida K & Hoffman RM Histone H3 lysine-trimethylation markers are decreased by recombinant methioninase and increased by methotrexate at concentrations which inhibit methionine-addicted osteosarcoma cell proliferation. Biochem Biophys Rep. 28 101177 2021. DOI: 10.1016/j.bbrep.2021.101177
29 Aoki Y Yamamoto J Tome Y Hamada K Masaki N Inubushi S Tashiro Y Bouvet M Endo I Nishida K & Hoffman RM Over-methylation of histone H3 lysines is a common molecular change among the three major types of soft-tissue sarcoma in patient-derived xenograft (PDX) mouse models. Cancer Genomics Proteomics. 18(6) 715 - 721 2021. DOI: 10.21873/cgp.20292
30 Hoffman RM Jacobsen SJ & Erbe RW Reversion to methionine independence in simian virus 40-transformed human and malignant rat fibroblasts is associated with altered ploidy and altered properties of transformation. Proc Natl Acad Sci USA. 76(3) 1313 - 1317 1979. DOI: 10.1073/pnas.76.3.1313
31 Hoffman RM Jacobsen SJ & Erbe RW Reversion to methionine independence by malignant rat and SV40-transformed human fibroblasts. Biochem Biophys Res Commun. 82(1) 228 - 234 1978. DOI: 10.1016/0006-291x(78)90600-9
32 Yamamoto J Aoki Y Han Q Sugisawa N Sun YU Hamada K Nishino H Inubushi S Miyake K Matsuyama R Bouvet M Endo I & Hoffman RM Reversion from methionine addiction to methionine independence results in loss of tumorigenic potential of highly-malignant lung-cancer cells. Anticancer Res. 41(2) 641 - 643 2021. DOI: 10.21873/anticanres.14815
33 Hoffman RM Development of recombinant methioninase to target the general cancer-specific metabolic defect of methionine dependence: a 40-year odyssey. Expert Opin Biol Ther. 15(1) 21 - 31 2015. DOI: 10.1517/14712598.2015.963050