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

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

MOTOKAZU SATO, QINGHONG HAN, YUTARO KUBOTA, ANTON BARANOV, DANIEL ARDJMAND, KOHEI MIZUTA, SEI MORINAGA, BYUNG MO KANG, NORITOSHI KOBAYASHI, MICHAEL BOUVET, YASUSHI ICHIKAWA, ATSUSHI NAKAJIMA and ROBERT M. HOFFMAN
Anticancer Research January 2024, 44 (1) 31-35; DOI: https://doi.org/10.21873/anticanres.16785
MOTOKAZU SATO
1AntiCancer Inc., San Diego, CA, U.S.A.;
2Department of Surgery, University of California, San Diego, CA, U.S.A.;
3Department of Oncology, Yokohama City University School of Medicine, Yokohama, Japan;
4Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
  • 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
YUTARO KUBOTA
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
ANTON BARANOV
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
DANIEL ARDJMAND
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
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
SEI MORINAGA
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
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
NORITOSHI KOBAYASHI
3Department of Oncology, Yokohama City University School of Medicine, Yokohama, Japan;
  • 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
YASUSHI ICHIKAWA
3Department of Oncology, Yokohama City University School of Medicine, Yokohama, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ATSUSHI NAKAJIMA
4Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
  • 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: Irinotecan (IRN), a topoisomerase I inhibitor and pro-drug of SN-38, is first-line treatment of colon cancer as part of FOLFIRI and FOLFOXIRI combination chemotherapy. However, IRN causes dose-limiting adverse events such as neutropenia and diarrhea. Dose reductions are sometimes required, which reduce efficacy. Recombinant methioninase (rMETase) targets the fundamental basis of cancer, methionine addiction, known as the Hoffman effect, and enhances the efficacy of numerous chemotherapy drugs. The present study determined the efficacy of rMETase when administered in combination with IRN. Materials and Methods: Cell viability was assessed by cultivating the HCT-116 human colorectal cancer cell line in 96-well plates at 1×103 cells per well in Dulbecco’s modified Eagle’s medium (DMEM). Subsequently, HCT-116 cells were treated with increasing concentrations of SN-38, the active form of IRN, ranging from 0.5 nM to 32 nM, and/or rMETase ranging from 0.125 to 8 U/ml. After treatment for 72 h, the half-maximal inhibitory concentration (IC50) of SN-38 alone and rMETase alone for HCT-116 cells were determined. Using the IC50 concentration of rMETase, we determined the IC50 of SN-38 in combination with rMETase. Cell viability was determined with the cell-counting Kit-8 with the WST-8 reagent.. Results: The IC50 of rMETase alone for the HCT-116 cells was 0.55 U/ml, and the IC50 of IRN (SN-38) alone was 3.50 nM. rMETase at 0.55 U/ml lowered the IC50 of SN-38 to 0.232 nM (p<0.0001), a 15-fold reduction. Conclusion: rMETase and IRN are strongly synergistic, giving rise to the possibility of lowering the effective dose of IRN for the treatment of patients with colon cancer, thereby reducing its severe toxicity. This new strategy will allow more patients with cancer to be effectively treated with IRN.

Key Words:
  • Colon cancer
  • irinotecan
  • toxicity
  • methionine
  • combination therapy
  • synergy
  • IC50
  • methionine addiction
  • Hoffman effect

Irinotecan (IRN) is a prodrug of SN-38 and exerts its efficacy by inhibiting topoisomerase I, which is required for DNA replication (1,2). IRN has been used as first- or second-line chemotherapy for over 20 years for a variety of cancers, including gastrointestinal, gynecological, and respiratory cancer (3). In particular, IRN has become first-line treatment for colorectal cancer with unresectable distant metastases (4). However, toxicity such as neutropenia and diarrhea, is a dose-limiting factor of IRN. Appropriate management of adverse effects, including dose reduction, is essential for the use of IRN, but reduces efficacy (3, 5).

All cancer cell types are addicted to methionine (6, 7) due at least partially to elevated levels of abnormal transmethylation (8-10). Methionine addiction, termed the Hoffman effect (9), is much stronger than the Warburg effect for glucose addiction (11). Methionine restriction (MR) has been observed to selectively arrest the cell cycle of cancer cells, specifically in the late-S/G2 phase (12, 13), a target of most chemotherapy drugs (14-17). However, methionine is present in many types of food and its restriction through dietary means alone is a challenge (18-21). Our laboratory has developed recombinant methioninase (rMETase), an enzyme that degrades methionine. Methioninase inhibits tumor growth in all malignancies in vitro and in vivo (22-27). Oral administration of rMETase (oral-rMETase) is effective in inhibiting tumor growth without side effects and has shown synergy with many chemotherapy drugs (27-35).

In the present study, we determined the synergistic efficacy of the combination of rMETase and SN-38, the active form of IRN, and whether the effective dose of IRN could be potentially reduced in order to eliminate severe side effects.

Materials and Methods

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

Recombinant methioninase (rMETase) production. Recombinant l-methionine α-deamino-γ-mercapto-methane lyase [recombinant methionase (rMETase)] (EC 4.4.1.11) from Pseudomonas putida was produced in recombinant Escherichia coli (AntiCancer, Inc., San Diego, CA, USA) as described previously (22, 34, 35).

Cell viability. Cell viability was assessed by cultivating HCT-116 cells at subconfluence overnight in 96-well plates, seeded at a density of 1.0×103 cells per well in DMEM. On the subsequent day, HCT-116 cells were treated with increasing concentrations of SN-38 ranging from 0.5 nM to 32 nM, and/or rMETase, increasing from 0.125 to 8 U/ml. SN-38 was obtained from MedChemExpress (Monmouth Junction, NJ, USA). Cell viability was determined after treatment for 72 h, using the Cell Counting Kit-8 (Dojindo Laboratory, Kumamoto, Japan) with the WST-8 reagent.

Determination of IC50 values. The software ImageJ version 1.54 (National Institutes of Health in Bethesda, MD, USA), was utilized for the computation of IC50 values and the construction of drug-sensitivity curves. Following the determination of the half-maximal inhibitory concentration (IC50) values for SN-38 alone and rMETase alone, the synergistic efficacy of their combination was assessed using the IC50 concentration of rMETase to determine its effects on the IC50 of SN-38. IC50 values were calculated using nonlinear regression.

Statistical analysis. Statistical significance was defined as p≤0.05. Statistical analyses were performed using GraphPad Prism 10.0.3 (GraphPad Software, Inc., San Diego, CA, USA).

Results

The IC50 of IRN (SN-38) alone was 3.50 nM and the IC50 of rMETase alone was 0.55 U/ml on HCT-116 cells (Figure 1 and Figure 2). Next, we investigated the efficacy of the combination of IRN (SN-38) and rMETase on HCT-116 cells in vitro. The reduction of the IC50 of IRN, in combination with the IC50 of rMETase, was 15-fold, from 3.50 nM to 0.232 nM (p<0.0001) (Figure 3).

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

Efficacy of SN-38 on HCT-116 cells in vitro. Cell viability was determined using the WST-8 reagent. The concentration axis uses a logarithmic base 2 scale. IC50: Half-maximal inhibitory concentration.

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

Efficacy of recombinant methioninase (rMETase) on HCT-116 cells in vitro. Cell viability was determined using the WST-8 reagent. The concentration axis uses a logarithmic base 2 scale. IC50: Half-maximal inhibitory concentration.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Recombinant methioninase (rMETase) lowered the half-maximal inhibitory concentration (IC50) of SN-38 on HCT-116 cells in vitro from 3.50 nM to 0.232 nM (p<0.001). Cell viability was determined using the WST-8 reagent. The concentration axis uses a logarithmic base 2 scale.

Discussion

The present study demonstrated a 15-fold reduction in the IC50 of IRN, by combining IRN with rMETase on HCT-116 colon cancer cells.

IRN (SN-38) binds and inhibits topoisomerase I, which plays a crucial role in DNA unwinding during replication and transcription. Inhibition of topoisomerase I leads to the arrest of the cell cycle in S-phase and the induction of apoptosis (36-38). The proximity of IRN (SN-38)’s action point to that of rMETase, which selectively arrests the cell cycle of cancer cells at the late S/G2 phase (12, 13), results in the very strong synergistic efficacy on HCT-116 colon cancer cells observed in the present study. rMETase acts synergistically with many other cytotoxic chemotherapy drugs (33).

A recent study showed greater efficacy of the combination of cytotoxic chemotherapy drugs with targeted drugs, as compared to cytotoxic–cytotoxic chemotherapy combinations. Particularly, one of the most effective combinations was between IRN (SN-38) and targeted agents, which act on the cell cycle (39).

Oral rMETase is showing promise in the clinic, especially in combination with chemotherapy (40-47).

Recently we have shown that rMETase greatly lowers the effective dose of regorafenib (48).

Conclusion

In the present study, the combination of IRN (SN-38) and rMETase had a synergistic effect, lowering the IC50 of SN-38 by 15-fold on colon cancer cells. The present study indicates the possibility of reducing the dose of IRN, without attenuating its efficacy, by combining it with rMETase, and provides an improved therapeutic opportunity for patients with colon cancer. rMETase is effective since it targets the fundamental and general hallmark of cancer, methionine addiction (6-10, 49-60).

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.

Footnotes

  • Authors’ Contributions

    Conception, design and experiments MS, YK and RMH. MS, KM and AB. MS and RMH wrote the article. QH provided methioninase. MS, YK, QH, AB, KM, SM, MB, NK, YI, AN, BMK and RMH critically reviewed the article.

  • Conflicts of Interest

    The Authors declare no competing interests regarding this work.

  • Received November 15, 2023.
  • Revision received December 4, 2023.
  • Accepted December 5, 2023.
  • Copyright © 2024 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

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. Hsiang YH,
    2. Liu LF
    : Identification of mammalian DNA topoisomerase I as an intracellular target of the anticancer drug camptothecin. Cancer Res 48: 1722-1726, 1988.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    1. Shao RG,
    2. Cao CX,
    3. Zhang H,
    4. Kohn KW,
    5. Wold MS,
    6. Pommier Y
    : Replication-mediated DNA damage by camptothecin induces phosphorylation of RPA by DNA-dependent protein kinase and dissociates RPA:DNA-PK complexes. EMBO J 18(5): 1397-1406, 1999. DOI: 10.1093/emboj/18.5.1397
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. de Man FM,
    2. Goey AKL,
    3. van Schaik RHN,
    4. Mathijssen RHJ,
    5. Bins S
    : Individualization of irinotecan treatment: a review of pharmacokinetics, pharmacodynamics, and pharmacogenetics. Clin Pharmacokinet 57(10): 1229-1254, 2018. DOI: 10.1007/s40262-018-0644-7
    OpenUrlCrossRefPubMed
  4. ↵
    1. Biller LH,
    2. Schrag D
    : Diagnosis and treatment of metastatic colorectal cancer. JAMA 325(7): 669, 2021. DOI: 10.1001/jama.2021.0106
    OpenUrlCrossRefPubMed
  5. ↵
    1. Kciuk M,
    2. Marciniak B,
    3. Kontek R
    : Irinotecan-still an important player in cancer chemotherapy: a comprehensive overview. Int J Mol Sci 21(14): 4919, 2020. DOI: 10.3390/ijms21144919
    OpenUrlCrossRefPubMed
  6. ↵
    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
  7. ↵
    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 USA 73(5): 1523-1527, 1976. DOI: 10.1073/pnas.73.5.1523
    OpenUrlAbstract/FREE Full Text
  8. ↵
    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
  9. ↵
    1. Kaiser P
    : Methionine dependence of cancer. Biomolecules 10(4): 568, 2020. DOI: 10.3390/biom10040568
    OpenUrlCrossRefPubMed
  10. ↵
    1. Wang Z,
    2. Yip LY,
    3. Lee JHJ,
    4. Wu Z,
    5. Chew HY,
    6. Chong PKW,
    7. Teo CC,
    8. Ang HY,
    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. Nat Med 25(5): 825-837, 2019. DOI: 10.1038/s41591-019-0423-5
    OpenUrlCrossRefPubMed
  11. ↵
    1. Kubota Y,
    2. Sato T,
    3. Hozumi C,
    4. Han Q,
    5. Aoki Y,
    6. Masaki N,
    7. Obara K,
    8. Tsunoda T,
    9. Hoffman RM
    : Superiority of [(11)C]methionine over [(18)F]deoxyglucose for PET imaging of multiple cancer types due to the methionine addiction of cancer. Int J Mol Sci 24(3): 1935, 2023. DOI: 10.3390/ijms24031935
    OpenUrlCrossRefPubMed
  12. ↵
    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
  13. ↵
    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
  14. ↵
    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. 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: 2157-2163, 1999.
    OpenUrlAbstract/FREE Full Text
    1. Yoshioka T,
    2. Wada T,
    3. Uchida N,
    4. Maki H,
    5. Yoshida H,
    6. Ide N,
    7. Kasai H,
    8. Hojo K,
    9. Shono K,
    10. Maekawa R,
    11. Yagi S,
    12. Hoffman RM,
    13. Sugita K
    : Anticancer efficacy in vivo and in vitro, synergy with 5-fluorouracil, and safety of recombinant methioninase. Cancer Res 58: 2583-2587, 1998.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Hoshiya Y,
    2. Kubota T,
    3. Inada T,
    4. Kitajima M,
    5. Hoffman RM
    : Methionine-depletion modulates the efficacy of 5-fluorouracil in human gastric cancer in nude mice. Anticancer Res 17: 4371-4375, 1997.
    OpenUrlPubMed
  16. ↵
    1. Epner DE
    : Can dietary methionine restriction increase the effectiveness of chemotherapy in treatment of advanced cancer? J Am Coll Nutr 20(5 Suppl): 443S-449S, 2001. DOI: 10.1080/07315724.2001.10719183
    OpenUrlCrossRefPubMed
    1. Epner DE,
    2. Morrow S,
    3. Wilcox M,
    4. Houghton JL
    : Nutrient intake and nutritional indexes in adults with metastatic cancer on a phase I clinical trial of dietary methionine restriction. Nutr Cancer 42(2): 158-166, 2002. DOI: 10.1207/S15327914NC422_2
    OpenUrlCrossRefPubMed
    1. Thivat E,
    2. Farges MC,
    3. Bacin F,
    4. D’Incan M,
    5. Mouret-Reynier MA,
    6. Cellarier E,
    7. Madelmont JC,
    8. Vasson MP,
    9. Chollet P,
    10. Durando X
    : Phase II trial of the association of a methionine-free diet with cystemustine therapy in melanoma and glioma. Anticancer Res 29: 5235-5240, 2009.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Durando X,
    2. Farges MC,
    3. Buc E,
    4. Abrial C,
    5. Petorin-Lesens C,
    6. Gillet B,
    7. Vasson MP,
    8. Pezet D,
    9. Chollet P,
    10. Thivat E
    : Dietary methionine restriction with FOLFOX regimen as first line therapy of metastatic colorectal cancer: a feasibility study. Oncology 78(3-4): 205-209, 2010. DOI: 10.1159/000313700
    OpenUrlCrossRefPubMed
  18. ↵
    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-mercapto-methane-lyase for novel anticancer therapy. Protein Expr Purif 9(2): 233-245, 1997. DOI: 10.1006/prep.1996.0700
    OpenUrlCrossRefPubMed
    1. Tan Y,
    2. Zavala J,
    3. Xu M,
    4. Zavala J and
    5. Hoffman RM
    : Serum methionine depletion without side effects by methioninase in metastatic breast cancer patients. Anticancer Res 16: 3937-3942, 1996.
    OpenUrlPubMed
    1. Igarashi K,
    2. Kawaguchi K,
    3. Kiyuna T,
    4. Miyake K,
    5. Murakami T,
    6. Yamamoto N,
    7. Hayashi K,
    8. Kimura H,
    9. Miwa S,
    10. Tsuchiya H,
    11. Hoffman RM
    : Effective metabolic targeting of human osteosarcoma cells in vitro and in orthotopic nude-mouse models with recombinant methioninase. Anticancer Res 37: 4807-4812, 2017. DOI: 10.21873/anticanres.11887
    OpenUrlAbstract/FREE Full Text
    1. Kawaguchi K,
    2. Igarashi K,
    3. Li S,
    4. Han Q,
    5. Tan Y,
    6. Kiyuna T,
    7. Miyake K,
    8. Murakami T,
    9. Chmielowski B,
    10. Nelson SD,
    11. Russell TA,
    12. Dry SM,
    13. Li Y,
    14. Unno M,
    15. Eilber FC,
    16. Hoffman RM
    : Combination treatment with recombinant methioninase enables temozolomide to arrest a BRAF V600E melanoma in a patient-derived orthotopic xenograft (PDOX) mouse model. Oncotarget 8(49): 85516-85525, 2017. DOI: 10.18632/oncotarget.20231
    OpenUrlCrossRefPubMed
    1. Miyake M,
    2. Miyake K,
    3. Han Q,
    4. Igarashi K,
    5. Kawaguchi K,
    6. Barangi M,
    7. Kiyuna T,
    8. Sugisawa N,
    9. Higuchi T,
    10. Oshiro H,
    11. Zhang Z,
    12. Razmjooei S,
    13. Bouvet M,
    14. Endo I,
    15. Hoffman RM
    : Synergy of oral recombinant methioninase (rMETase) and 5-fluorouracil on poorly differentiated gastric cancer. Biochem Biophys Res Commun 643: 48-54, 2023. DOI: 10.1016/j.bbrc.2022.12.062.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Miyake K,
    2. Kiyuna T,
    3. Li S,
    4. Han Q,
    5. Tan Y,
    6. Zhao M,
    7. Oshiro H,
    8. Kawaguchi K,
    9. Higuchi T,
    10. Zhang Z,
    11. Razmjooei S,
    12. Barangi M,
    13. Wangsiricharoen S,
    14. Murakami T,
    15. Singh AS,
    16. Li Y,
    17. Nelson SD,
    18. Eilber FC,
    19. Bouvet M,
    20. Hiroshima Y,
    21. Chishima T,
    22. Matsuyama R,
    23. Singh SR,
    24. Endo I,
    25. Hoffman RM
    : Combining tumor-selective bacterial therapy with Salmonella typhimurium A1-R and cancer metabolism targeting with oral recombinant methioninase regressed an Ewing’s sarcoma in a patient-derived orthotopic xenograft model. Chemotherapy 63(5): 278-283, 2018. DOI: 10.1159/000495574
    OpenUrlCrossRefPubMed
    1. Aoki Y,
    2. Tome Y,
    3. Han Q,
    4. Yamamoto J,
    5. Hamada K,
    6. Masaki N,
    7. Kubota Y,
    8. Bouvet M,
    9. Nishida K,
    10. Hoffman RM
    : Oral-recombinant methioninase converts an osteosarcoma from methotrexate-resistant to -sensitive in a patient-derived orthotopic-xenograft (PDOX) mouse model. Anticancer Res 42(2): 731-737, 2022. DOI: 10.21873/anticanres.15531
    OpenUrlAbstract/FREE Full Text
    1. Hoffman RM,
    2. Han Q,
    3. Kawaguchi K,
    4. Li S,
    5. Tan Y
    : Afterword: Oral methioninase–answer to cancer and fountain of youth? Methods Mol Biol 1866: 311-322, 2019. DOI: 10.1007/978-1-4939-8796-2_24
    OpenUrlCrossRefPubMed
    1. Oshiro H,
    2. Tome Y,
    3. Kiyuna T,
    4. Yoon SN,
    5. Lwin TM,
    6. Han Q,
    7. Tan Y,
    8. Miyake K,
    9. Higuchi T,
    10. Sugisawa N,
    11. Katsuya Y,
    12. Park JH,
    13. Zang Z,
    14. Razmjooei S,
    15. Bouvet M,
    16. Clary B,
    17. Singh SR,
    18. Kanaya F,
    19. Nishida K,
    20. Hoffman RM
    : Oral recombinant methioninase overcomes colorectal-cancer liver metastasis resistance to the combination of 5-Fluorouracil and oxaliplatinum in a patient-derived orthotopic xenograft mouse model. Anticancer Res 39(9): 4667-4671, 2019. DOI: 10.21873/anticanres.13648
    OpenUrlAbstract/FREE Full Text
    1. Park JH,
    2. Zhao M,
    3. Han Q,
    4. Sun Y,
    5. Higuchi T,
    6. Sugisawa N,
    7. Yamamoto J,
    8. Singh SR,
    9. Clary B,
    10. Bouvet M,
    11. Hoffman RM
    : Efficacy of oral recombinant methioninase combined with oxaliplatinum and 5-fluorouracil on primary colon cancer in a patient-derived orthotopic xenograft mouse model. Biochem Biophys Res Commun 518(2): 306-310, 2019. DOI: 10.1016/j.bbrc.2019.08.051
    OpenUrlCrossRefPubMed
    1. Yamamoto J,
    2. Miyake K,
    3. Han Q,
    4. Tan Y,
    5. Inubushi S,
    6. Sugisawa N,
    7. Higuchi T,
    8. Tashiro Y,
    9. Nishino H,
    10. Homma Y,
    11. Matsuyama R,
    12. Chawla SP,
    13. Bouvet M,
    14. Singh SR,
    15. Endo I,
    16. Hoffman RM
    : Oral recombinant methioninase increases TRAIL receptor-2 expression to regress pancreatic cancer in combination with agonist tigatuzumab in an orthotopic mouse model. Cancer Lett 492: 174-184, 2020. DOI: 10.1016/j.canlet.2020.07.034
    OpenUrlCrossRefPubMed
  20. ↵
    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
  21. ↵
    1. Hoffman RM
    1. Hoffman RM,
    2. Tan Y,
    3. Li S,
    4. Han Q,
    5. Yagi S,
    6. Takakura T,
    7. Takimoto A,
    8. Inagaki K,
    9. Kudou D
    : Development of recombinant methioninase for cancer treatment. In: Methionine Dependence of Cancer and Aging: Methods and Protocols. Hoffman RM (ed.). New York, NY, Springer, pp. 107-131, 2019.
  22. ↵
    1. 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
    OpenUrlCrossRefPubMed
  23. ↵
    1. Kaku Y,
    2. Tsuchiya A,
    3. Kanno T,
    4. Nishizaki T
    : Irinotecan induces cell cycle arrest, but not apoptosis or necrosis, in Caco-2 and CW2 colorectal cancer cell lines. Pharmacology 95(3-4): 154-159, 2015. DOI: 10.1159/000381029
    OpenUrlCrossRefPubMed
    1. Magrini R,
    2. Bhonde MR,
    3. Hanski ML,
    4. Notter M,
    5. Scherübl H,
    6. Boland CR,
    7. Zeitz M,
    8. Hanski C
    : Cellular effects of CPT 11 on colon carcinoma cells: Dependence on p53 and hMLH1 status. Int J Cancer 101(1): 23-31, 2002. DOI: 10.1002/ijc.10565
    OpenUrlCrossRefPubMed
  24. ↵
    1. Bhonde MR,
    2. Hanski ML,
    3. Notter M,
    4. Gillissen BF,
    5. Daniel PT,
    6. Zeitz M,
    7. Hanski C
    : Equivalent effect of DNA damage-induced apoptotic cell death or long-term cell cycle arrest on colon carcinoma cell proliferation and tumour growth. Oncogene 25(2): 165-175, 2006. DOI: 10.1038/sj.onc.1209017
    OpenUrlCrossRefPubMed
  25. ↵
    1. Jaaks P,
    2. Coker EA,
    3. Vis DJ,
    4. Edwards O,
    5. Carpenter EF,
    6. Leto SM,
    7. Dwane L,
    8. Sassi F,
    9. Lightfoot H,
    10. Barthorpe S,
    11. van der Meer D,
    12. Yang W,
    13. Beck A,
    14. Mironenko T,
    15. Hall C,
    16. Hall J,
    17. Mali I,
    18. Richardson L,
    19. Tolley C,
    20. Morris J,
    21. Thomas F,
    22. Lleshi E,
    23. Aben N,
    24. Benes CH,
    25. Bertotti A,
    26. Trusolino L,
    27. Wessels L,
    28. Garnett MJ
    : Effective drug combinations in breast, colon and pancreatic cancer cells. Nature 603(7899): 166-173, 2022. DOI: 10.1038/s41586-022-04437-2
    OpenUrlCrossRefPubMed
  26. ↵
    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
    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
    1. Kubota Y,
    2. Han Q,
    3. Hozumi C,
    4. Masaki N,
    5. Yamamoto J,
    6. Aoki Y,
    7. Tsunoda T,
    8. Hoffman RM
    : Stage IV pancreatic cancer patient treated with FOLFIRINOX combined with oral methioninase: a highly-rare case with long-term stable disease. Anticancer Res 42(5): 2567-2572, 2022. DOI:10.21873/anticanres.15734
    OpenUrlAbstract/FREE Full Text
    1. Yamamoto J,
    2. Han Q,
    3. Simon M,
    4. Thomas D,
    5. Hoffman RM
    : Methionine restriction: Ready for prime time in the cancer clinic? Anticancer Res 42(2): 641-644, 2022. DOI:10.21873/anticanres.15521
    OpenUrlAbstract/FREE Full Text
    1. Kubota Y,
    2. Han Q,
    3. Hamada K,
    4. Aoki Y,
    5. Masaki N,
    6. Obara K,
    7. Tsunoda T,
    8. Hoffman RM
    : Long-term Stable Disease in a Rectal-cancer Patient Treated by Methionine Restriction With Oral Recombinant Methioninase and a Low-methionine Diet. Anticancer Res 42(8): 3857-3861, 2022. DOI: 10.21873/anticanres.15877
    OpenUrlAbstract/FREE Full Text
    1. Han Q,
    2. Hoffman RM
    : Chronic treatment of an advanced prostate-cancer patient with oral methioninase resulted in long-term stabilization of rapidly rising PSA levels. In Vivo 35(4): 2171-2176, 2021. DOI: 10.21873/invivo.12488
    OpenUrlAbstract/FREE Full Text
    1. Han Q,
    2. Hoffman RM
    : Lowering and stabilizing PSA levels in advanced-prostate cancer patients with oral methioninase. Anticancer Res 41(4): 1921-1926, 2021. DOI: 10.21873/anticanres.14958
    OpenUrlAbstract/FREE Full Text
  27. ↵
    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
  28. ↵
    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. Aoki Y,
    2. Kubota Y,
    3. Han Q,
    4. Masaki N,
    5. Obara K,
    6. Bouvet M,
    7. Chawla SP,
    8. Tome Y,
    9. Nishida K,
    10. Hoffman RM.
    The combination of methioninase and ethionine exploits methionine addiction to selectively eradicate osteosarcoma cells and not normal cells and synergistically down-regulates the expression of C-MYC. Cancer Genomics Proteomics 20(6suppl): 679-685, 2023. DOI: 10.21873/cgp.20415
    OpenUrlAbstract/FREE Full Text
    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
    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
    1. Yamamoto J,
    2. Aoki Y,
    3. Inubushi S,
    4. Han Q,
    5. Hamada K,
    6. Tashiro Y,
    7. Miyake K,
    8. Matsuyama R,
    9. Bouvet M,
    10. Clarke SG,
    11. Endo I,
    12. 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
    OpenUrlAbstract/FREE Full Text
    1. Yamamoto J,
    2. Aoki Y,
    3. Han Q,
    4. Sugisawa N,
    5. Sun YU,
    6. Hamada K,
    7. Nishino H,
    8. Inubushi S,
    9. Miyake K,
    10. Matsuyama R,
    11. Bouvet M,
    12. Endo I,
    13. 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
    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. 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 76(3): 1313, 1979. DOI: 10.1073/pnas.76.3.1313
    OpenUrlAbstract/FREE Full Text
    1. Hoffman RM,
    2. Jacobsen SJ,
    3. 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
    OpenUrlCrossRefPubMed
    1. Pokrovsky VS,
    2. Qoura LA,
    3. Demidova EA,
    4. Han Q,
    5. Hoffman RM
    : Targeting methionine addiction of cancer cells with methioninase. Biochemistry (Mosc) 88(7): 944-952, 2023. DOI: 10.1134/S0006297923070076
    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. Biochem Biophys Res Commun 117(2): 429-34, 1983. DOI: 10.1016/0006-291x(83)91218-4
    OpenUrlCrossRefPubMed
    1. Tan Y,
    2. Xu M,
    3. Hoffman RM
    : Broad selective efficacy of recombinant methioninase and polyethylene glycol-modified recombinant methioninase on cancer cells In Vitro. Anticancer Res 30(4): 1041-6, 2010.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Stern PH,
    2. Wallace CD,
    3. Hoffman RM
    : Altered methionine metabolism occurs in all members of a set of diverse human tumor cell lines. J Cell Physiol 119(1): 29-34, 1984. DOI: 10.1002/jcp.1041190106
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 44 (1)
Anticancer Research
Vol. 44, Issue 1
January 2024
  • 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.
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
(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.
10 + 10 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
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
MOTOKAZU SATO, QINGHONG HAN, YUTARO KUBOTA, ANTON BARANOV, DANIEL ARDJMAND, KOHEI MIZUTA, SEI MORINAGA, BYUNG MO KANG, NORITOSHI KOBAYASHI, MICHAEL BOUVET, YASUSHI ICHIKAWA, ATSUSHI NAKAJIMA, ROBERT M. HOFFMAN
Anticancer Research Jan 2024, 44 (1) 31-35; DOI: 10.21873/anticanres.16785

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
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
MOTOKAZU SATO, QINGHONG HAN, YUTARO KUBOTA, ANTON BARANOV, DANIEL ARDJMAND, KOHEI MIZUTA, SEI MORINAGA, BYUNG MO KANG, NORITOSHI KOBAYASHI, MICHAEL BOUVET, YASUSHI ICHIKAWA, ATSUSHI NAKAJIMA, ROBERT M. HOFFMAN
Anticancer Research Jan 2024, 44 (1) 31-35; DOI: 10.21873/anticanres.16785
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • 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...

  • Human fibrosarcoma cells selected for very-high doxorubicin resistance, acquire trabectedin and eribulin cross-resistance, remain sensitive to recombinant methioninase, and have increased c-MYC expression
  • Recombinant Methioninase and Cisplatinum Act Synergistically to Inhibit Lewis Lung Carcinoma Cells But Not Normal Fibroblasts
  • Synergistic Eradication of Fibrosarcoma With Acquired Ifosfamide Resistance Using Methionine Restriction Combined With Ifosfamide in Nude-mouse Models
  • Ivermectin Combined With Recombinant Methioninase (rMETase) Synergistically Eradicates MiaPaCa-2 Pancreatic Cancer Cells
  • Selective Synergy of Recombinant Methioninase Plus Docetaxel Against Docetaxel-resistant and -sensitive Fibrosarcoma Cells Compared to Normal Fibroblasts
  • Recombinant Methioninase Synergistically Reverses High-docetaxel Resistance Developed in Osteosarcoma Cells
  • Extensive DNA Damage and Loss of Cell Viability Occur Synergistically With the Combination of Recombinant Methioninase and Paclitaxel on Pancreatic Cancer Cells which Report DNA-Damage Response in Real Time
  • Therapeutic Responses to Two New SN-38 Derivatives in Colorectal Cancer Patient-Derived Xenografts and Respective 3D In Vitro Cultures
  • Overcoming High Trabectedin Resistance of Soft-tissue Sarcoma With Recombinant Methioninase: A Potential Solution of a Recalcitrant Clinical Problem
  • Recombinant Methioninase Increases Eribulin Efficacy 16-fold in Highly Eribulin-resistant HT1080 Fibrosarcoma Cells, Demonstrating Potential to Overcome the Clinical Challenge of Drug-resistant Soft-tissue Sarcoma
  • [11C]Methionine PET vs. [18F]Fluorodeoxyglucose PET Whole-body Imaging to Determine the Extent of Methionine-addiction Compared to Glucose-addiction of Primary and Metastatic Cancer of the Trunk in Patients
  • Recombinant Methioninase Is Selectively Synergistic With Doxorubicin Against Wild-type Fibrosarcoma Cells Compared to Normal Cells and Overcomes Highly-Doxorubicin-resistant Fibrosarcoma
  • DNA-Binding Agent Trabectedin Combined With Recombinant Methioninase Is Synergistic to Decrease Fibrosarcoma Cell Viability and Induce Nuclear Fragmentation But Not Synergistic on Normal Fibroblasts
  • Targeting Methionine Addiction Combined With Low-dose Irinotecan Arrested Colon Cancer in Contrast to High-dose Irinotecan Alone, Which Was Ineffective, in a Nude-mouse Model
  • Reduction of Tumor Biomarkers from very High to Normal and Extensive Metastatic Lesions to Undetectability in a Patient With Stage IV HER2-positive Breast Cancer Treated With Low-dose Trastuzumab Deruxtecan in Combination With Oral Recombinant Methioninase and a Low-methionine Diet
  • Google Scholar

More in this TOC Section

  • Tumor Immunology Visualized in Realtime in In-vivo-like 3-D Gelfoam® Histoculture by Color-coded Imaging
  • The Role of EMP2 in Triple-negative Breast Cancer
  • Role of the Circadian Clock Gene DEC1 in Chemosensitivity and Apoptosis in Colorectal Cancer
Show more Experimental Studies

Similar Articles

Keywords

  • colon cancer
  • irinotecan
  • toxicity
  • methionine
  • combination therapy
  • synergy
  • IC50
  • methionine addiction
  • Hoffman effect
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire