Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies
Open Access

The Triple Combination of Recombinant Methioninase, Rapamycin, and Chloroquine Synergistically Eradicates HCT116 Colon Cancer Cells

JINSOO KIM, QINGHONG HAN, BYUNG MO KANG, KOHEI MIZUTA, YOHEI ASANO, MICHAEL BOUVET and ROBERT M. HOFFMAN
Anticancer Research July 2025, 45 (7) 2773-2779; DOI: https://doi.org/10.21873/anticanres.17646
JINSOO KIM
1AntiCancer Inc., San Diego, CA, U.S.A.
2Department of Surgery, University of California, San Diego, CA, U.S.A.
3Department of Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
QINGHONG HAN
1AntiCancer Inc., San Diego, CA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
BYUNG MO KANG
1AntiCancer Inc., San Diego, CA, U.S.A.
2Department of Surgery, University of California, San Diego, CA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KOHEI MIZUTA
1AntiCancer Inc., San Diego, CA, U.S.A.
2Department of Surgery, University of California, San Diego, CA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YOHEI ASANO
1AntiCancer Inc., San Diego, CA, U.S.A.
2Department of Surgery, University of California, San Diego, CA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MICHAEL BOUVET
2Department of Surgery, University of California, San Diego, CA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ROBERT M. HOFFMAN
1AntiCancer Inc., San Diego, CA, U.S.A.
2Department of Surgery, University of California, San Diego, CA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: all@anticancer.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: The prognosis for advanced/metastatic colon cancer is poor, with low survival rates despite aggressive treatment. Recombinant methioninase (rMETase) targets the methionine addiction of cancer and works synergistically with many anticancer drugs. The present study aimed to determine the synergistic efficacy of the combination of rMETase, rapamycin (RAPA), and chloroquine (CQ) on human HCT116 colon cancer cells in vitro.

Materials and Methods: The half-maximal inhibitory concentrations (IC50) of rMETase, RAPA, and CQ were determined for the human HCT116 cell line in vitro. The synergy of rMETase combined with RAPA; rMETase combined with CQ; and rMETase combined with RAPA and CQ, at their respective IC50 values, was determined on HCT116 cells. Cell viability was measured with the WST-8 reagent.

Results: The IC50 value of rMETase was 0.48 U/ml; for RAPA 19.6 μM; and for CQ 16.5 μM on the HCT116 cell line. Synergy was observed with both the combination of rMETase and RAPA (p=0.02) and the combination of rMETase and CQ (p=0.03). The triple combination of rMETase, RAPA, plus CQ showed the strongest synergistic efficacy, eradicating HCT116 cells (p=0.01).

Conclusion: The triple combination of rMETase, RAPA and CQ shows strong synergistic efficacy on the human HCT116 colon cancer cell line. The results of the present study suggest the potential for future clinical applications of this triple-combination treatment for advanced/metastatic colon cancer.

Keywords:
  • Methioninase
  • rapamycin
  • chloroquine
  • synergy
  • HCT116 cells
  • colon cancer
  • methionine addiction
  • Hoffman effect

Introduction

In 1959, Sugimura et al. demonstrated that tumor growth was inhibited in rat models when methionine was excluded from the diet (1). Subsequently, it was observed that cancer cells synthesize normal or greater-than-normal levels of methionine despite their absolute requirement for exogenous methionine (2). These findings led to the concept of methionine addiction in cancer cells, known as the Hoffman effect (3-21). Methionine addiction is due to the excessive utilization of methionine for transmethylation reactions within cancer cells, resulting in their dependency on exogenous methionine (11-17).

Methionine restriction inhibits DNA synthesis by inducing cell-cycle arrest at the late S/G2 phase in cancer cells (18, 19). An effective approach to limiting methionine availability in cancer cells involves the use of methioninase, an enzyme which degrades methionine (20). Our previous investigations have demonstrated the synergistic efficacy of recombinant methioninase (rMETase) in combination with numerous chemotherapeutic agents and targeted therapies in multiple cancer types (22). The selective cell-cycle arrest by rMETase of cancer cells in the S/G2 phase of the cell cycle increases the efficacy of cancer drugs, which also target S/G2.

The mammalian target of rapamycin (mTOR) pathway regulates cell proliferation and protein synthesis in cancer cells (23). Rapamycin (RAPA) is an mTOR inhibitor and has demonstrated clinical efficacy on renal-cell carcinoma and breast cancer (24). We have demonstrated that rMETase and RAPA were synergistic on cancer cells in vitro and in vivo (25-26).

Autophagy degrades damaged organelles and recycles intracellular components to sustain metabolic balance (27). Chloroquine (CQ) inhibits autophagy, leading to apoptosis in cancer cells due to autophagosome accumulation (27, 28). Although CQ is not used as a monotherapy in cancer treatment due to the high dose required for efficacy, its combination with standard therapeutic regimens has been proposed to overcome cancer resistance (29, 30).

Water-soluble derivatives of RAPA (e.g., temsirolimus and everolimus) and CQ (e.g., hydroxychloroquine) had synergistic efficacy on solid tumors and melanoma in phase I clinical trials (31). We have previously demonstrated the synergy of RAPA and CQ in liposarcoma using patient-derived orthotopic xenograft (PDOX) mouse models and in vitro systems (32, 33). We recently demonstrated the synergistic efficacy of the triple combination of rMETase, RAPA, and CQ on osteosarcoma cells in vitro (34).

Building on our previous findings, the present study aimed to investigate whether the triple combination of rMETase, RAPA, and CQ has synergistic efficacy on the HCT116 colon cancer cell line.

Materials and Methods

Cell culture. The HCT-116 human colon carcinoma cell line was obtained from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured in Dulbecco’s Modified Eagle’s Medium/Nutrient Mixture F-12 with GlutaMAX™ supplement (DMEM/F-12), along with with 10% fetal bovine serum (FBS) and 100 IU/ml penicillin/streptomycin, at 37°C in a 5% CO2 incubator.

Recombinant methioninase production. rMETase was produced by AntiCancer Inc. (San Diego, CA, USA). The methioninase gene, cloned from Pseudomonas putida, was introduced into Escherichia coli and expressed using fermentation. rMETase purification involved a 60°C thermal step, polyethylene glycol precipitation, and diethylaminoethyl (DEAE)-sepharose ion-exchange chromatography, as previously described (35).

Reagents. RAPA and CQ were obtained from MedChem-Express (Monmouth Junction, NJ, USA) and dissolved in dimethyl sulfoxide (DMSO) at a stock concentration of 10 mM.

Cell viability assay. Cell viability was assessed using the WST-8 assay (Dojindo Laboratories, Kumamoto, Japan). HCT-116 cells were seeded at 2.0×103 cells/well in 96-well plates and incubated overnight. Cells were treated with varying concentrations of rMETase (0.125-8 U/ml), RAPA, or CQ (1-128 μM) for 72 h. After treatment, 10 μl of WST-8 solution was added to each well, and absorbance was measured at 450 nm using a microplate reader (Sunrise; Tecan, Männedorf, Switzerland). Drug sensitivity curves and half-maximal inhibitory concentration (IC50) values were generated using Microsoft 365 Excel for MacOS (Microsoft, Redmond, WA, USA), ImageJ ver. 1.54g (National Institutes of Health, Bethesda, MD, USA), and GraphPad Prism ver. 10.4.1 (GraphPad Software, Inc., San Diego, CA, USA).

Combination treatment. HCT116 cells were seeded in 96-well plates at a density of 2.0×103 cells/well in DMEM/F-12. After 24 h, the cells were treated as follows: 1) control (DMEM/F-12 only); 2) rMETase at its IC50 concentration (0.48 U/m); 3) RAPA at its IC50 concentration (19.6 μM); 4) CQ at its IC50 concentration (16.5 μM); 5) rMETase (0.48 U/ml) plus RAPA (19.6 μM); 6) rMETase (0.48 U/ml) plus CQ (16.5 μM); and 7) rMETase (0.48 U/ml) plus RAPA (19.6 μM) plus CQ (16.5 μM). After 72 h treatment, cell viability was determined with the WST-8 reagent. Experiments were performed in triplicate.

Statistical analysis. Data are presented as mean±standard deviation. Groups were compared using one-way analysis of variance (ANOVA). Statistical significance was evaluated using Tukey’s multiple comparison test, with p-values ≤0.05 considered significant.

Results

Determination of IC50 values of rMETase, RAPA, and CQ on HCT116 cells. The IC50 value of rMETase was 0.48 U/ml; RAPA was 19.6 μM; and CQ was 16.5 μM for HCT116 cells (Figure 1).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Determination of the half-maximal inhibitory concentration (IC50) of recombinant methioninase (rMETase), rapamycin (RAPA), and chloroquine (CQ) on HCT116 cells in vitro. Cell viability was measured using the WST-8 assay. Data are shown as the mean±standard deviation. Please see the Materials and Methods for details.

Synergy of rMETase, RAPA, and CQ on HCT116 cells. All agents alone significantly reduced the viability of the HCT116 cells compared to the control cells: rMETase alone (0.48 U/ml, IC50) reduced the viability of HCT116 cells by approximately 37.5% (p=0.03). RAPA alone (19.6 μM, IC50) reduced the viability of HCT116 cells by 52.4.0% (p=0.04). CQ alone (16.5 μM, IC50) reduced the viability of HCT116 cells by 42.1% (p=0.05). rMETase plus RAPA reduced the viability of cells by 82.2% (p= 0.02). rMETase plus CQ reduced the viability of HCT116 cells by 66.7% (p=0.03). rMETase plus RAPA plus CQ essentially eradicated HCT116 cells by 95.2% (p= 0.01). The triple combination of rMETase, RAPA and CQ caused significantly greater reduction in the viability of HCT116 cells compared to monotherapies or the dual combinations rMETase plus RAPA or rMETase plus CQ (p< 0.001; Figure 2).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Synergy of the combination of recombinant methioninase (rMETase), rapamycin (RAPA), and chloroquine (CQ) at their IC50 values on HCT116 colon cancer cells. Data are shown as the mean±standard deviation. Please see the Materials and Methods for details. Significantly different at: *p<0.05, **p<0.01, ***p<0.001.

Discussion

Colorectal cancer is the third most common malignancy and the second leading cause of cancer-related mortality worldwide (36). Although surgery, chemotherapy, and radiation have improved the prognosis of localized colorectal cancer with an approximately 65% 5-year overall survival rate, the survival rate for metastatic/advanced disease is only approximately 14% (37). Therefore, new treatment strategies are needed for the treatment of metastatic colorectal cancer (38).

Based on our previous demonstrations of synergy of rMETase and chemotherapy drugs in colon cancer in vitro and in mouse models (39-42), the present study demonstrated that the triple combination of rMETase, RAPA, and CQ was highly synergistic in HCT116 colon cancer cells. Methionine restriction reduces the intracellular S-adenosylmethionine (SAM) concentration, which in turn inhibits SAMTOR binding and mTOR activity (6, 43). This may explain the synergy of rMETase and RAPA. Methionine restriction also affects autophagy, which may explain the synergy of rMETase and CQ (44, 45). We recently reported the synergy of rMETase, RAPA, and CQ on the 143B osteosarcoma cell line (34). The present study shows that HCT116 cells are more sensitive to the double combinations of rMETase plus RAPA and rMETase plus CQ than 143B osteosarcoma cells determined previously (34).

The triple combination of rMETase, RAPA, and CQ will be tested on colon-cancer mouse models and in the clinic in the future.

Conclusion

The triple synergistic effect of rMETase, RAPA, and CQ on HCT116 colon cancer cells can be readily translated to the clinic as RAPA and CQ are approved drugs and rMETase is given to cancer patients as a dietary enzyme supplement (46, 47, 48).

rMETase is effective as it targets the fundamental hallmark of cancer, methionine addiction (2-21).

Acknowledgements

This article is dedicated to the memory of A.R. Moossa, MD, Professor Philip Miles, Sun Lee, MD, Richard W. Erbe, MD, Professor Milton Plesur, Professor Gordon H. Sato, John W. Littlefield, MD, Professor Li Jiaxi, Masaki Kitajima, MD, Shigeo Yagi, PhD, Jack Geller, MD, Joseph R. Bertino, MD, J.A.R. Mead, PhD, Eugene P. Frenkel, MD, John Medelsohn, MD, Professor Lev Bergelson, Professor Sheldon Penman, Professor John R. Raper and Joseph Leighton, MD.

The Robert M. Hoffman Foundation for Cancer Research provided funds for the present study.

Footnotes

  • Authors’ Contributions

    JK and RMH designed the study. QH produced rMETase. JK conducted all experiments and wrote the article. RMH revised the article. BMK, KM, YA, and MB critically read the manuscript.

  • Conflicts of Interest

    The Authors have no conflicts of interest or financial ties to disclose related to this study.

  • Artificial Intelligence (AI) Disclosure

    No artificial intelligence (AI) tools, including large language models or machine learning software, were used in the preparation, analysis, or presentation of this manuscript.

  • Received April 8, 2025.
  • Revision received April 28, 2025.
  • Accepted April 29, 2025.
  • Copyright © 2025 The Author(s). Published by the International Institute of Anticancer Research.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Sugimura T,
    2. Birnbaum SM,
    3. Winitz M,
    4. Greenstein JP
    : Quantitative nutritional studies with water-soluble, chemically defined diets. IX. Further studies on d-glucosamine-containing diets. Arch Biochem Biophys 83(2): 521-527, 1959. DOI: 10.1016/0003-9861(59)90060-8
    OpenUrlCrossRefPubMed
  2. ↵
    1. Hoffman RM,
    2. Erbe RW
    : High in vivo rates of methionine biosynthesis in transformed human and malignant rat cells auxotrophic for methionine. Proc Natl Acad Sci U S A 73(5): 1523-1527, 1976. DOI: 10.1073/pnas.73.5.1523
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Halpern BC,
    2. Clark BR,
    3. Hardy DN,
    4. Halpern RM,
    5. Smith RA
    : The effect of replacement of methionine by homocystine on survival of malignant and normal adult mammalian cells in culture. Proc Natl Acad Sci U S A 71(4): 1133-1136, 1974. DOI: 10.1073/pnas.71.4.1133
    OpenUrlAbstract/FREE Full Text
    1. Hoffman RM,
    2. Jacobsen SJ,
    3. 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
    OpenUrlAbstract/FREE Full Text
    1. Hoffman RM
    : Altered methionine metabolism, DNA methylation and oncogene expression in carcinogenesis. A review and synthesis. Biochim Biophys Acta 738(1-2): 49-87, 1984. DOI: 10.1016/0304-419x(84)90019-2
    OpenUrlCrossRefPubMed
  4. ↵
    1. Coalson DW,
    2. Mecham JO,
    3. Stern PH,
    4. 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
    OpenUrlAbstract/FREE Full Text
    1. Stern PH,
    2. Mecham JO,
    3. Wallace CD,
    4. 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
    OpenUrlCrossRefPubMed
    1. Stern PH,
    2. 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): 629-639, 1986. DOI: 10.1093/jnci/76.4.629
    OpenUrlCrossRefPubMed
    1. Mecham JO,
    2. Rowitch D,
    3. Wallace CD,
    4. Stern PH,
    5. Hoffman RM
    : The metabolic defect of methionine dependence occurs frequently in human tumor cell lines. Biochemical and Biophysical Research Communications 117(2): 429-434, 1983. DOI: 10.1016/0006-291X(83)91218-4
    OpenUrlCrossRefPubMed
    1. Yamamoto J,
    2. Inubushi S,
    3. Han Q,
    4. Tashiro Y,
    5. Sugisawa N,
    6. Hamada K,
    7. Aoki Y,
    8. Miyake K,
    9. Matsuyama R,
    10. Bouvet M,
    11. Clarke SG,
    12. Endo I,
    13. Hoffman RM
    : Linkage of methionine addiction, histone lysine hypermethylation, and malignancy. iScience 25(4): 104162, 2022. DOI: 10.1016/j.isci.2022.104162
    OpenUrlCrossRefPubMed
  5. ↵
    1. Stern PH,
    2. 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
    OpenUrlCrossRefPubMed
    1. Wang Z,
    2. Yip LY,
    3. Lee JHJ,
    4. Wu Z,
    5. Chew HY,
    6. Chong PKW,
    7. Teo CC,
    8. Ang HY-K,
    9. Peh KLE,
    10. Yuan J,
    11. Ma S,
    12. Choo LSK,
    13. Basri N,
    14. Jiang X,
    15. Yu Q,
    16. Hillmer AM,
    17. Lim WT,
    18. Lim TKH,
    19. Takano A,
    20. Tan EH,
    21. Tan DSW,
    22. Ho YS,
    23. Lim B,
    24. Tam WL
    : Methionine is a metabolic dependency of tumor-initiating cells. Nature Medicine 25(5): 825-837, 2019. DOI: 10.1038/s41591-019-0423-5
    OpenUrlCrossRefPubMed
    1. Tisdale MJ
    : Effect of methionine deprivation on methylation and synthesis of macromolecules. Br J Cancer 42(1): 121-128, 1980. DOI: 10.1038/bjc.1980.210
    OpenUrlCrossRefPubMed
    1. Judde JG,
    2. Ellis M,
    3. Frost P
    : Biochemical analysis of the role of transmethylation in the methionine dependence of tumor cells. Cancer Res 49(17): 4859-4865, 1989.
    OpenUrlAbstract/FREE Full Text
    1. Yamamoto J,
    2. Han Q,
    3. Inubushi S,
    4. Sugisawa N,
    5. Hamada K,
    6. Nishino H,
    7. Miyake K,
    8. Kumamoto T,
    9. Matsuyama R,
    10. Bouvet M,
    11. Endo I,
    12. 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
    OpenUrlCrossRefPubMed
    1. Aoki Y,
    2. Han Q,
    3. Tome Y,
    4. Yamamoto J,
    5. Kubota Y,
    6. Masaki N,
    7. Obara K,
    8. Hamada K,
    9. Wang JD,
    10. Inubushi S,
    11. Bouvet M,
    12. Clarke SG,
    13. Nishida K,
    14. 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
    OpenUrlCrossRefPubMed
  6. ↵
    1. Montalbano S,
    2. Raboni S,
    3. Sidoli S,
    4. Mozzarelli A,
    5. Bettati S,
    6. Buschini A
    : Post-translational modifications of histone variants in the absence and presence of a methionine-depleting enzyme in normal and cancer cells. Cancers (Basel) 15(2): 527, 2023. DOI: 10.3390/cancers15020527
    OpenUrlCrossRefPubMed
  7. ↵
    1. Hoffman RM,
    2. Jacobsen SJ
    : Reversible growth arrest in simian virus 40-transformed human fibroblasts. Proc Natl Acad Sci USA 77(12): 7306-7310, 1980. DOI: 10.1073/pnas.77.12.7306
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Yano S,
    2. Li S,
    3. Han Q,
    4. Tan Y,
    5. Bouvet M,
    6. Fujiwara T,
    7. 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
    OpenUrlCrossRefPubMed
  9. ↵
    1. Abo Qoura L,
    2. Balakin KV,
    3. Hoffman RM,
    4. Pokrovsky VS
    : The potential of methioninase for cancer treatment. Biochim Biophys Acta Rev Cancer 1879(4): 189122, 2024. DOI: 10.1016/j.bbcan.2024.189122
    OpenUrlCrossRef
  10. ↵
    1. Lin DW,
    2. Carranza FG,
    3. Borrego S,
    4. Lauinger L,
    5. Dantas de Paula L,
    6. Pulipelli HR,
    7. Andronicos A,
    8. Hertel KJ,
    9. Kaiser P
    : Nutrient control of splice site selection contributes to methionine addiction of cancer. Mol Metab 93: 102103, 2025. DOI: 10.1016/j.molmet.2025.102103
    OpenUrlCrossRefPubMed
  11. ↵
    1. Kubota Y,
    2. Han Q,
    3. Aoki Y,
    4. Masaki N,
    5. Obara K,
    6. Hamada K,
    7. Hozumi C,
    8. Wong ACW,
    9. Bouvet M,
    10. Tsunoda T,
    11. 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
    OpenUrlCrossRefPubMed
  12. ↵
    1. Vignot S,
    2. Faivre S,
    3. Aguirre D,
    4. Raymond E
    : mTOR-targeted therapy of cancer with rapamycin derivatives. Ann Oncol 16(4): 525-537, 2005. DOI: 10.1093/annonc/mdi113
    OpenUrlCrossRefPubMed
  13. ↵
    1. Raymond E,
    2. Alexandre J,
    3. Faivre S,
    4. Vera K,
    5. Materman E,
    6. Boni J,
    7. Leister C,
    8. Korth-Bradley J,
    9. Hanauske A,
    10. Armand JP
    : Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 22(12): 2336-2347, 2004. DOI: 10.1200/JCO.2004.08.116
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Masaki N,
    2. Han Q,
    3. Samonte C,
    4. Wu NF,
    5. Hozumi C,
    6. Wu J,
    7. Obara K,
    8. Kubota Y,
    9. Aoki Y,
    10. Bouvet M,
    11. Hoffman RM
    : Oral-recombinant methioninase in combination with rapamycin eradicates osteosarcoma of the breast in a patient-derived orthotopic xenograft mouse model. Anticancer Res 42(11): 5217-5222, 2022. DOI: 10.21873/anticanres.16028
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Ardjmand D,
    2. Kubota Y,
    3. Sato M,
    4. Han Q,
    5. Mizuta K,
    6. Morinaga S,
    7. Hoffman RM
    : Selective synergy of rapamycin combined with methioninase on cancer cells compared to normal cells. Anticancer Res 44(3): 929-933, 2024. DOI: 10.21873/anticanres.16887
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. White E
    : The role for autophagy in cancer. J Clin Invest 125(1): 42-46, 2015. DOI: 10.1172/JCI73941
    OpenUrlCrossRefPubMed
  17. ↵
    1. Button RW,
    2. Roberts SL,
    3. Willis TL,
    4. Hanemann CO,
    5. Luo S
    : Accumulation of autophagosomes confers cytotoxicity. J Biol Chem 292(33): 13599-13614, 2017. DOI: 10.1074/jbc.M117.782276
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Pascolo S
    : Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies. Eur J Pharmacol 771: 139-144, 2016. DOI: 10.1016/j.ejphar.2015.12.017
    OpenUrlCrossRefPubMed
  19. ↵
    1. Hassan AMIA,
    2. Zhao Y,
    3. Chen X,
    4. He C
    : Blockage of autophagy for cancer therapy: a comprehensive review. Int J Mol Sci 25(13): 7459, 2024. DOI: 10.3390/ijms25137459
    OpenUrlCrossRefPubMed
  20. ↵
    1. Rangwala R,
    2. Chang YC,
    3. Hu J,
    4. Algazy KM,
    5. Evans TL,
    6. Fecher LA,
    7. Schuchter LM,
    8. Torigian DA,
    9. Panosian JT,
    10. Troxel AB,
    11. Tan KS,
    12. Heitjan DF,
    13. DeMichele AM,
    14. Vaughn DJ,
    15. Redlinger M,
    16. Alavi A,
    17. Kaiser J,
    18. Pontiggia L,
    19. Davis LE,
    20. O’Dwyer PJ,
    21. Amaravadi RK
    : Combined MTOR and autophagy inhibition: phase I trial of hydroxychloroquine and temsirolimus in patients with advanced solid tumors and melanoma. Autophagy 10(8): 1391-1402, 2014. DOI: 10.4161/auto.29119
    OpenUrlCrossRefPubMed
  21. ↵
    1. Masaki N,
    2. Aoki Y,
    3. Kubota Y,
    4. Obara K,
    5. Miyazaki J,
    6. Hoffman RM
    : Chloroquine combined with rapamycin arrests tumor growth in a patient-derived orthotopic xenograft (PDOX) mouse model of dedifferentiated liposarcoma. In Vivo 36(6): 2630-2637, 2022. DOI: 10.21873/invivo.12997
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Masaki N,
    2. Aoki Y,
    3. Obara K,
    4. Kubota Y,
    5. Bouvet M,
    6. Miyazaki J,
    7. Hoffman RM
    : Targeting autophagy with the synergistic combination of chloroquine and rapamycin as a novel effective treatment for well-differentiated liposarcoma. Cancer Genomics Proteomics 20(4): 317-322, 2023. DOI: 10.21873/cgp.20384
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Bouvet M,
    6. Yamamoto N,
    7. Hayashi K,
    8. Kimura H,
    9. Miwa S,
    10. Igarashi K,
    11. Higuchi T,
    12. Tsuchiya H,
    13. Demura S,
    14. Hoffman RM
    : Highly synergistic eradication of 143B osteosarcoma cells in vitro by the combination of recombinant methioninase, chloroquine, and rapamycin targeting methionine addiction, autophagy, and mTOR, respectively. Anticancer Res 45(3): 935-941, 2025. DOI: 10.21873/anticanres.17481
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Tan Y,
    2. Xu M,
    3. Tan X,
    4. Tan X,
    5. Wang X,
    6. Saikawa Y,
    7. Nagahama T,
    8. Sun X,
    9. Lenz M,
    10. 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
    OpenUrlCrossRefPubMed
  25. ↵
    1. Siegel RL,
    2. Giaquinto AN,
    3. Jemal A
    : Cancer statistics, 2024. CA Cancer J Clin 74(1): 12-49, 2024. DOI: 10.3322/caac.21820
    OpenUrlCrossRefPubMed
  26. ↵
    1. Siegel RL,
    2. Wagle NS,
    3. Cercek A,
    4. Smith RA,
    5. Jemal A
    : Colorectal cancer statistics, 2023. CA Cancer J Clin 73(3): 233-254, 2023. DOI: 10.3322/caac.21772
    OpenUrlCrossRefPubMed
  27. ↵
    1. Kuipers EJ,
    2. Grady WM,
    3. Lieberman D,
    4. Seufferlein T,
    5. Sung JJ,
    6. Boelens PG,
    7. van de Velde CJ,
    8. Watanabe T
    : Colorectal cancer. Nat Rev Dis Primers 1: 15065, 2015. DOI: 10.1038/nrdp.2015.65
    OpenUrlCrossRefPubMed
  28. ↵
    1. Tan Y,
    2. Sun X,
    3. Xu M,
    4. Tan X,
    5. Sasson A,
    6. Rashidi B,
    7. Han Q,
    8. Tan X,
    9. Wang X,
    10. An Z,
    11. Sun FX,
    12. Hoffman RM
    : Efficacy of recombinant methioninase in combination with cisplatin on human colon tumors in nude mice. Clin Cancer Res 5(8): 2157-2163, 1999.
    OpenUrlAbstract/FREE Full Text
    1. Park JH,
    2. Han Q,
    3. Zhao M,
    4. Tan Y,
    5. Higuchi T,
    6. Yoon SN,
    7. Sugisawa N,
    8. Yamamoto J,
    9. Bouvet M,
    10. Clary B,
    11. Singh SR,
    12. Hoffman RM
    : Oral recombinant methioninase combined with oxaliplatinum and 5-fluorouracil regressed a colon cancer growing on the peritoneal surface in a patient-derived orthotopic xenograft mouse model. Tissue Cell 61: 109-114, 2019. DOI: 10.1016/j.tice.2019.09.006
    OpenUrlCrossRefPubMed
    1. Choobin BB,
    2. Kubota Y,
    3. Han Q,
    4. Ardjmand D,
    5. Morinaga S,
    6. Mizuta K,
    7. Bouvet M,
    8. Tsunoda T,
    9. Hoffman RM
    : Recombinant methioninase lowers the effective dose of regorafenib against colon-cancer cells: a strategy for widespread clinical use of a toxic drug. Cancer Diagn Progn 3(6): 655-659, 2023. DOI: 10.21873/cdp.10268
    OpenUrlCrossRefPubMed
  29. ↵
    1. Sato M,
    2. Han Q,
    3. Kubota Y,
    4. Baranov A,
    5. Ardjmand D,
    6. Mizuta K,
    7. Morinaga S,
    8. Kang BM,
    9. Kobayashi N,
    10. Bouvet M,
    11. Ichikawa Y,
    12. Nakajima A,
    13. Hoffman RM
    : Recombinant methioninase decreased the effective dose of irinotecan by 15-fold against colon cancer cells: a strategy for effective low-toxicity treatment of colon cancer. Anticancer Res 44(1): 31-35, 2024. DOI: 10.21873/anticanres.16785
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Gu X,
    2. Orozco JM,
    3. Saxton RA,
    4. Condon KJ,
    5. Liu GY,
    6. Krawczyk PA,
    7. Scaria SM,
    8. Harper JW,
    9. Gygi SP,
    10. Sabatini DM
    : SAMTOR is an S-adenosylmethionine sensor for the mTORC1 pathway. Science 358(6364): 813-818, 2017. DOI: 10.1126/science.aao3265
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Xin L,
    2. Zhou LQ,
    3. Liu L,
    4. Yuan YW,
    5. Zhang HT,
    6. Zeng F
    : METase promotes cell autophagy via promoting SNHG5 and suppressing miR-20a in gastric cancer. Int J Biol Macromol 122: 1046-1052, 2019. DOI: 10.1016/j.ijbiomac.2018.09.051
    OpenUrlCrossRefPubMed
  32. ↵
    1. Xin L,
    2. Li SH,
    3. Liu C,
    4. Zeng F,
    5. Cao JQ,
    6. Zhou LQ,
    7. Zhou Q,
    8. Yuan YW
    : Methionine represses the autophagy of gastric cancer stem cells via promoting the methylation and phosphorylation of RAB37. Cell Cycle 19(20): 2644-2652, 2020. DOI: 10.1080/15384101.2020.1814044
    OpenUrlCrossRefPubMed
  33. ↵
    1. Han Q,
    2. Tan Y,
    3. Hoffman RM
    : Oral dosing of recombinant methioninase is associated with a 70% drop in PSA in a patient with bone-metastatic prostate cancer and 50% reduction in circulating methionine in a high-stage ovarian cancer patient. Anticancer Res 40(5): 2813-2819, 2020. DOI: 10.21873/anticanres.14254
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Kubota Y,
    2. Han Q,
    3. Masaki N,
    4. Hozumi C,
    5. Hamada K,
    6. Aoki Y,
    7. Obara K,
    8. Tsunoda T,
    9. Hoffman RM
    : Elimination of axillary-lymph-node metastases in a patient with invasive lobular breast cancer treated by first-line neo-adjuvant chemotherapy combined with methionine restriction. Anticancer Res 42(12): 5819-5823, 2022. DOI: 10.21873/anticanres.16089
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Kubota Y,
    2. Han Q,
    3. Morinaga S,
    4. Tsunoda T,
    5. Hoffman RM
    : Rapid reduction of CEA and stable metastasis in an NRAS-mutant rectal-cancer patient treated with FOLFIRI and bevacizumab combined with oral recombinant methioninase and a low-methionine diet upon metastatic recurrence after FOLFIRI and bevacizumab treatment alone. In Vivo 37(5): 2134-2138, 2023. DOI: 10.21873/invivo.13310
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

Anticancer Research: 45 (7)
Anticancer Research
Vol. 45, Issue 7
July 2025
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
The Triple Combination of Recombinant Methioninase, Rapamycin, and Chloroquine Synergistically Eradicates HCT116 Colon Cancer Cells
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
4 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
The Triple Combination of Recombinant Methioninase, Rapamycin, and Chloroquine Synergistically Eradicates HCT116 Colon Cancer Cells
JINSOO KIM, QINGHONG HAN, BYUNG MO KANG, KOHEI MIZUTA, YOHEI ASANO, MICHAEL BOUVET, ROBERT M. HOFFMAN
Anticancer Research Jul 2025, 45 (7) 2773-2779; DOI: 10.21873/anticanres.17646

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
The Triple Combination of Recombinant Methioninase, Rapamycin, and Chloroquine Synergistically Eradicates HCT116 Colon Cancer Cells
JINSOO KIM, QINGHONG HAN, BYUNG MO KANG, KOHEI MIZUTA, YOHEI ASANO, MICHAEL BOUVET, ROBERT M. HOFFMAN
Anticancer Research Jul 2025, 45 (7) 2773-2779; DOI: 10.21873/anticanres.17646
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Conclusion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Magnolol Suppresses Osteosarcoma Progression via Apoptosis Induction and EGFR/AKT Pathway Inactivation in a U-2 OS Xenograft Model
  • The Combination of the Autophagy Inhibitor Chloroquine and Recombinant Methioninase Has Selective Synergistic Efficacy on Human Colon Cancer Cells But Not on Normal Human Fibroblasts
  • Amentoflavone Enhances the Anti-tumor Activity of Regorafenib by Promoting Apoptosis and Inhibiting NF-κB–mediated Metastasis in Hepatocellular Carcinoma
Show more Experimental Studies

Similar Articles

Keywords

  • Methioninase
  • rapamycin
  • Chloroquine
  • synergy
  • HCT116 cells
  • colon cancer
  • methionine addiction
  • Hoffman effect
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire