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

Selective Synergy of Ivermectin Combined With Recombinant Methioninase Against Colon-Cancer Cells in Contrast to Normal Fibroblasts

YOHEI ASANO, QINGHONG HAN, SHUKUAN LI, KOHEI MIZUTA, BYUNG MO KANG, JIN SOO KIM, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, SEI MORINAGA, HIROYUKI TSUCHIYA, SATORU DEMURA and ROBERT M. HOFFMAN
Anticancer Research June 2025, 45 (6) 2257-2263; DOI: https://doi.org/10.21873/anticanres.17600
YOHEI ASANO
1AntiCancer Inc., San Diego, CA, U.S.A.;
2Department of Surgery, University of California, San Diego, CA, U.S.A.;
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 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
SHUKUAN LI
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
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
JIN SOO KIM
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
NORIO YAMAMOTO
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KATSUHIRO HAYASHI
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROAKI KIMURA
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHINJI MIWA
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KENTARO IGARASHI
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TAKASHI HIGUCHI
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SEI MORINAGA
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROYUKI TSUCHIYA
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SATORU DEMURA
3Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 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: Recent studies have shown that ivermectin, developed as an anti-parasitic drug, has efficacy against several cancer types. Methionine restriction, including the use of recombinant methioninase (rMETase), has been developed to target methionine addiction, a fundamental hallmark of cancer, termed the Hoffman effect. Metastatic colorectal cancer (CRC) is a recalcitrant disease that requires novel and disruptive treatment approaches. The present study aimed to determine the efficacy of ivermectin in combination with rMETase on a CRC cell line compared to normal fibroblasts.

Materials and Methods: The human CRC cell line HCT-116 and normal human fibroblasts Hs27 were seeded at a density of 1,000 cells per well in 96-well plates and cultured overnight at 37°C. After treatment with ivermectin (0.5 μM to 128 μM) or rMETase (0.0625 U/ml to 8 U/ml) for 72 h, cell viability was assessed using the WST-8 reagent to determine the half-maximal inhibitory concentration (IC50) values for ivermectin and rMETase on both cell lines. Using these IC50 values, cell-viability assays for ivermectin alone, rMETase alone, and the combination of ivermectin and rMETase were performed on both cell lines to determine their synergy.

Results: The IC50 value for ivermectin alone was 4.81 μM and rMETase alone was 0.61 U/ml against HCT-116 colon-cancer cells. The IC50 value for ivermectin alone was 8.67 μM and rMETase alone was 0.67 U/ml against Hs27 normal fibroblasts. In HCT-116 cells, treatment with the combination of ivermectin and rMETase resulted in greater reduction of cell proliferation, compared to treatment with each drug alone; however, no synergy was observed against Hs27 cells.

Conclusion: rMETase and ivermectin showed selective synergistic anti-cancer efficacy against colon-cancer cells, indicating the clinical potential of the combination for metastatic CRC.

Keywords:
  • Ivermectin
  • recombinant methioninase (rMETase)
  • combination
  • synergy
  • colorectal cancer
  • methionine addiction
  • Hoffman effect
  • normal fibroblasts

Introduction

Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide, with over 1.9 million new cases and approximately 935,000 deaths reported in 2022 (1). Targeted therapy and immunotherapy have been used for CRC (2,3), but metastatic CRC (mCRC) is still a recalcitrant disease due to frequent resistance to standard treatment, which limits the efficacy of current therapeutic strategies.

Ivermectin belongs to the class of 16-membered macrocyclic lactones and has been widely used as an anti-parasitic agent (4-8). It has been shown to induce apoptosis to inhibit cancer-cell proliferation in breast, lung, colon, and biliary cancers (4, 9-13). Ivermectin also inhibits cancer-cell proliferation by inducing autophagy mediated by p21-activated kinase (PAK1) as well as caspase-dependent apoptosis (14).

Our laboratory has developed recombinant methioninase (rMETase) to target methionine addiction in cancer, a phenomenon known as the Hoffman effect (15-19). Previous studies have demonstrated the therapeutic efficacy of methionine restriction, including rMETase on CRC (20-23). Furthermore, we have shown that the combination of rMETase and ivermectin was synergistic on breast and pancreatic cancer cells in vitro (24, 25).

In the present study, we focused on the combination of rMETase and ivermectin as a novel treatment strategy for CRC. The aim of the present study was to determine the synergy of rMETase and ivermectin on a human CRC cell line in vitro compared to normal fibroblasts.

Materials and Methods

Cell culture. The human HCT-116 colon-cancer cell line and Hs27 normal human fibroblasts were obtained from the American Type Culture Collection (Manassas, VA, USA). These cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (ThermoFisher Scientific, Waltham, MA, USA) at 37°C in a 5% CO2 incubator.

Reagents. rMETase used in the present study was produced at AntiCancer Inc. (San Diego, CA, USA) by fermentation of recombinant Escherichia coli transformed with the Pseudomonas putida methioninase gene as described previously (26). Ivermectin was obtained from MedChemoExpress (Monmouth Junction, NJ, USA).

Cell viability assay. Cell viability assays were performed using the WST-8 reagent (Dojindo Laboratory, Kumamoto, Japan) to determine the half-maximal inhibitory concentration (IC50) of ivermectin and rMETase on the HCT-116 and Hs27 cell lines. Both cell lines were seeded in 100 μl DMEM at 1,000 cells/well in 96-well plates and incubated overnight. After confirming the proliferation of these cells, they were treated with rMETase at concentrations ranging from 0.0625 U/ml to 8 U/ml, or with ivermectin at concentrations ranging from 0.5 μM to 128 μM for 72 h. Then, 10 μl of WST-8 solution was added to each well. After 1 h, the absorbance at 450 nm was measured using a microplate reader (Sunrise, Tecan, Mannedorf, Switzerland). Drug-sensitivity curves were generated using Microsoft Excel for Mac 2024 ver. 16.89.1 (Microsoft, Redmond, WA, USA), ImageJ ver. 1.54g (National Institutes of Health, Bethesda, MD, USA), and GraphPad Prism 10.4.1 (GraphPad Software, Inc., San Diego, CA, USA). The IC50 was then calculated. These experiments were performed twice, each in triplicate.

Synergy determination of the combination of rMETase and ivermectin. To evaluate the synergy of the combination of rMETase and ivermectin, HCT-116 and Hs27 cells were seeded in 96-well plates and assigned to one of four treatment groups: (i) control (DMEM); (ii) rMETase (IC50); (iii) ivermectin (IC50); or (iv) the combination of rMETase and ivermectin (each at IC50). After 72 h treatment, cell viability was assessed using the WST-8 assay, and absorbance was measured to determine cell viability.

Statistical analysis. All experiments in the present study were performed twice, each in triplicate. All data are presented as mean±standard deviation. Comparisons between treatment groups were calculated using one-way analysis of variance (ANOVA), followed by Tukey’s multiple comparison test. Statistical analyses were performed using EZR software (Saitama Medical Center, Jichi Medical University, Saitama, Japan), with p≤0.05 considered statistically significant.

Results

Determination of the IC50 of ivermectin and rMETase on HCT-116 and Hs27 cells. Drug sensitivity curves of HCT-116 and Hs27 treated with rMETase and ivermectin were generated (Figure 1, Figure 2). For HCT-116 colon-cancer cells, the IC50 value of rMETase was 0.61 U/ml and the IC50 of ivermectin was 4.81 μM. For normal Hs27 fibroblasts, the IC50 values of rMETase and ivermectin, were 0.67 U/ml and 8.67 μM, respectively.

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

Drug-sensitivity curves and half-maximal inhibitory concentrations (IC50) for (A) ivermectin and (B) recombinant methioninase (rMETase) on HCT-116 colon-cancer cells. Please see the Materials and Methods for details.

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

Half-maximal inhibitory concentration (IC50) for (A) ivermectin and (B) recombinant methioninase (rMETase) on Hs27 normal human fibroblasts. Please see the Materials and Methods for details.

Determination of synergy of the combination of rMETase and ivermectin on HCT-116 and Hs27 cells. The IC50 concentrations determined from the drug-sensitivity curves were used to assess viability of HCT-116 and Hs27 cells treated with rMETase alone; ivermectin alone; or a combination of rMETase and ivermectin. The combination resulted in significantly lower cell viability of the HCT-116 cell line, compared to rMETase and ivermectin monotherapy (both p<0.05, Figure 3A). In contrast, although rMETase alone and ivermectin alone inhibited Hs27 cells, their combination did not show enhanced efficacy (Figure 3B).

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

Synergy determination of ivermectin and recombinant methioninase (rMETase) on (A) HCT-116 colon-cancer cells and (B) Hs27 normal human fibroblasts treated with ivermectin (IC50) alone, rMETase (IC50) alone, and their combination. Please see the Materials and Methods for details.

Discussion

rMETase targets methionine addiction, a fundamental hallmark of cancer cells (15-21, 27-37). Methionine restriction selectively arrests cancer cells at the late S/G2 phase of the cell cycle (35, 36). rMETase has been demonstrated to enhance the efficacy of numerous chemotherapy drugs that target cells in S-phase (18, 20-22, 38-41).

Ivermectin belongs to the class of 16-membered macrocyclic lactones and has been widely used as an anti-parasitic agent (4-8). Ivermectin has been shown to induce caspase-dependent apoptosis in cancer cells and to inhibit cancer-cell proliferation by autophagy via PAK1 (4, 9-14). The present study demonstrated promising selective and synergistic efficacy of the combination of ivermectin and rMETase against colorectal-cancer cells (HCT-116) but not normal human fibroblasts (Hs27). The present results suggest a critical difference between cancer cells and normal cells, possibly related to the methionine addiction of cancer cells.

Ivermectin and methionine restriction, including rMETase, inhibit the S-phase of the cell cycle of cancer cells (11, 35, 36, 42, 43), which may account for their synergy. Previous in vitro studies have shown the efficacy of ivermectin against gemcitabine-resistant cholangiocarcinoma cells and paclitaxel-resistant non-small cell lung cancer cells (11, 43), as well as breast and lung cancer (9, 10, 12, 13). However, few studies have been conducted on the combination of ivermectin with other drugs to determine anti-cancer efficacy. The present study is the first to show that ivermectin in combination with rMETase may have synergistic efficacy on colorectal-cancer cells.

Our previous in vitro studies have reported the synergistic efficacy of rMETase and ivermectin on breast and pancreatic cancers (24, 25). As mentioned above, ivermectin can induce autophagy and apoptosis in cancer cells (4, 9-14). Future experiments will investigate the efficacy of rMETase combined with ivermectin on apoptosis and autophagy of cancer cells compared to normal cells.

Limitations of the study include its in vitro design and the use of a single cancer cell line. In addition, the present study is focused on the effect or rMETase and ivermectin on cell proliferation, leaving uninvestigated the underlying mechanisms of these effects. Further studies that will be performed on a larger number of in vitro and in vivo models of colorectal cancer, are necessary to confirm our promising findings.

Conclusion

The combination of ivermectin and rMETase enhanced the anti-cancer efficacy of these agents against colon-cancer cells. The synergy was selective for cancer cells. The results of the present study can be readily translated to the clinic for recalcitrant cancer, as ivermectin is an FDA approved drug and rMETase is used as a dietary enzyme supplement for cancer patients (38, 39).

Acknowledgements

This article is dedicated to the memory of A.R. Moossa, MD, Sun Lee, MD, Professor Philip Miles, Richard W. Erbe, MD, Professor Milton Plesur, Professor Gordon H. Sato, John W. Littlefield, MD, Professor Li Jiaxi, Masaki Kitajima, MD, Joseph R. Bertino, MD, Shigeo Yagi, PhD, J.A.R Mead, PhD. Eugene P. Frenkel, MD, John Mendelsohn, 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

    YA and RMH designed the study. QH provided rMETase. YA performed experiments and formal analysis. YA was the major contributor to writing – original draft and RMH revised the manuscript. QH, KM, BMK, JSK, NY, KH, HK, ShM, KI, TH, SeM, HT, and SD critically read and approved the final manuscript.

  • Conflicts of Interest

    The Authors have no conflicts of interest.

  • 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 2, 2025.
  • Revision received May 5, 2025.
  • Accepted May 12, 2025.
  • Copyright © 2025 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. Bray F,
    2. Laversanne M,
    3. Sung H,
    4. Ferlay J,
    5. Siegel RL,
    6. Soerjomataram I,
    7. Jemal A
    : Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 74(3): 229-263, 2024. DOI: 10.3322/caac.21834
    OpenUrlCrossRef
  2. ↵
    1. Van Cutsem E,
    2. Cervantes A,
    3. Adam R,
    4. Sobrero A,
    5. Van Krieken JH,
    6. Aderka D,
    7. Aranda Aguilar E,
    8. Bardelli A,
    9. Benson A,
    10. Bodoky G,
    11. Ciardiello F,
    12. D’Hoore A,
    13. Diaz-Rubio E,
    14. Douillard JY,
    15. Ducreux M,
    16. Falcone A,
    17. Grothey A,
    18. Gruenberger T,
    19. Haustermans K,
    20. Heinemann V,
    21. Hoff P,
    22. Köhne CH,
    23. Labianca R,
    24. Laurent-Puig P,
    25. Ma B,
    26. Maughan T,
    27. Muro K,
    28. Normanno N,
    29. Österlund P,
    30. Oyen WJ,
    31. Papamichael D,
    32. Pentheroudakis G,
    33. Pfeiffer P,
    34. Price TJ,
    35. Punt C,
    36. Ricke J,
    37. Roth A,
    38. Salazar R,
    39. Scheithauer W,
    40. Schmoll HJ,
    41. Tabernero J,
    42. Taïeb J,
    43. Tejpar S,
    44. Wasan H,
    45. Yoshino T,
    46. Zaanan A,
    47. Arnold D
    : ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 27(8): 1386-1422, 2016. DOI: 10.1093/annonc/mdw235
    OpenUrlCrossRefPubMed
  3. ↵
    1. Le DT,
    2. Durham JN,
    3. Smith KN,
    4. Wang H,
    5. Bartlett BR,
    6. Aulakh LK,
    7. Lu S,
    8. Kemberling H,
    9. Wilt C,
    10. Luber BS,
    11. Wong F,
    12. Azad NS,
    13. Rucki AA,
    14. Laheru D,
    15. Donehower R,
    16. Zaheer A,
    17. Fisher GA,
    18. Crocenzi TS,
    19. Lee JJ,
    20. Greten TF,
    21. Duffy AG,
    22. Ciombor KK,
    23. Eyring AD,
    24. Lam BH,
    25. Joe A,
    26. Kang SP,
    27. Holdhoff M,
    28. Danilova L,
    29. Cope L,
    30. Meyer C,
    31. Zhou S,
    32. Goldberg RM,
    33. Armstrong DK,
    34. Bever KM,
    35. Fader AN,
    36. Taube J,
    37. Housseau F,
    38. Spetzler D,
    39. Xiao N,
    40. Pardoll DM,
    41. Papadopoulos N,
    42. Kinzler KW,
    43. Eshleman JR,
    44. Vogelstein B,
    45. Anders RA,
    46. Diaz LA Jr.
    : Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357(6349): 409-413, 2017. DOI: 10.1126/science.aan6733
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Juarez M,
    2. Schcolnik-Cabrera A,
    3. Dueñas-Gonzalez A
    : The multitargeted drug ivermectin: From an antiparasitic agent to a repositioned cancer drug. Am J Cancer Res 8(2): 317-331, 2018.
    OpenUrlPubMed
    1. Anselmi M,
    2. Buonfrate D,
    3. Guevara Espinoza A,
    4. Prandi R,
    5. Marquez M,
    6. Gobbo M,
    7. Montresor A,
    8. Albonico M,
    9. Racines Orbe M,
    10. Martin Moreira J,
    11. Bisoffi Z
    : Mass administration of ivermectin for the elimination of onchocerciasis significantly reduced and maintained low the prevalence of Strongyloides stercoralis in Esmeraldas, Ecuador. PLoS Negl Trop Dis 9(11): e0004150, 2015. DOI: 10.1371/journal.pntd.0004150
    OpenUrlCrossRefPubMed
    1. King CL,
    2. Suamani J,
    3. Sanuku N,
    4. Cheng YC,
    5. Satofan S,
    6. Mancuso B,
    7. Goss CW,
    8. Robinson LJ,
    9. Siba PM,
    10. Weil GJ,
    11. Kazura JW
    : A trial of a triple-drug treatment for lymphatic filariasis. N Engl J Med 379(19): 1801-1810, 2018. DOI: 10.1056/NEJMoa1706854
    OpenUrlCrossRefPubMed
    1. Pinilla YT,
    2. C P Lopes S,
    3. S Sampaio V,
    4. Andrade FS,
    5. Melo GC,
    6. Orfanó AS,
    7. Secundino NFC,
    8. Guerra MGVB,
    9. Lacerda MVG,
    10. Kobylinski KC,
    11. Escobedo-Vargas KS,
    12. López-Sifuentes VM,
    13. Stoops CA,
    14. Baldeviano GC,
    15. Tarning J,
    16. Vasquez GM,
    17. Pimenta PFP,
    18. Monteiro WM
    : Promising approach to reducing Malaria transmission by ivermectin: Sporontocidal effect against Plasmodium vivax in the South American vectors Anopheles aquasalis and Anopheles darlingi. PLoS Negl Trop Dis 12(2): e0006221, 2018. DOI: 10.1371/journal.pntd.0006221
    OpenUrlCrossRefPubMed
  5. ↵
    1. Van Voorhis WC,
    2. Hooft van Huijsduijnen R,
    3. Wells TN
    : Profile of William C. Campbell, Satoshi Ōmura, and Youyou Tu, 2015 Nobel Laureates in Physiology or Medicine. Proc Natl Acad Sci USA 112(52): 15773-15776, 2015. DOI: 10.1073/pnas.1520952112
    OpenUrlFREE Full Text
  6. ↵
    1. Melotti A,
    2. Mas C,
    3. Kuciak M,
    4. Lorente-Trigos A,
    5. Borges I,
    6. Ruiz i Altaba A
    : The river blindness drug Ivermectin and related macrocyclic lactones inhibit WNT-TCF pathway responses in human cancer. EMBO Mol Med 6(10): 1263-1278, 2014. DOI: 10.15252/emmm.201404084
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Dou Q,
    2. Chen HN,
    3. Wang K,
    4. Yuan K,
    5. Lei Y,
    6. Li K,
    7. Lan J,
    8. Chen Y,
    9. Huang Z,
    10. Xie N,
    11. Zhang L,
    12. Xiang R,
    13. Nice EC,
    14. Wei Y,
    15. Huang C
    : Ivermectin induces cytostatic autophagy by blocking the PAK1/Akt axis in breast cancer. Cancer Res 76(15): 4457-4469, 2016. DOI: 10.1158/0008-5472.Can-15-2887
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Intuyod K,
    2. Hahnvajanawong C,
    3. Pinlaor P,
    4. Pinlaor S
    : Anti-parasitic drug ivermectin exhibits potent anticancer activity against gemcitabine-resistant cholangiocarcinoma in vitro. Anticancer Res 39(9): 4837-4843, 2019. DOI: 10.21873/anticanres.13669
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Wang K,
    2. Gao W,
    3. Dou Q,
    4. Chen H,
    5. Li Q,
    6. Nice EC,
    7. Huang C
    : Ivermectin induces PAK1-mediated cytostatic autophagy in breast cancer. Autophagy 12(12): 2498-2499, 2016. DOI: 10.1080/15548627.2016.1231494
    OpenUrlCrossRefPubMed
  10. ↵
    1. Dominguez-Gomez G,
    2. Chavez-Blanco A,
    3. Medina-Franco JL,
    4. Saldivar-Gonzalez F,
    5. Flores-Torrontegui Y,
    6. Juarez M,
    7. Díaz-Chávez J,
    8. Gonzalez-Fierro A,
    9. Dueñas-González A
    : Ivermectin as an inhibitor of cancer stem-like cells. Mol Med Rep 17(2): 3397-3403, 2018. DOI: 10.3892/mmr.2017.8231
    OpenUrlCrossRef
  11. ↵
    1. Liu J,
    2. Zhang K,
    3. Cheng L,
    4. Zhu H,
    5. Xu T
    : Progress in understanding the molecular mechanisms underlying the antitumour effects of ivermectin. Drug Des Devel Ther 14: 285-296, 2020. DOI: 10.2147/DDDT.S237393
    OpenUrlCrossRefPubMed
  12. ↵
    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
    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 U S A 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
  13. ↵
    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
  14. ↵
    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
  15. ↵
    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
  16. ↵
    1. Kim MJ,
    2. Han Q,
    3. Bouvet M,
    4. Hoffman RM,
    5. Park JH
    : Recombinant oral methioninase (o-rMETase) combined with oxaliplatinum plus 5-fluorouracil improves survival of mice with massive colon-cancer peritoneal carcinomatosis. Anticancer Res 43(1): 19-24, 2023. DOI: 10.21873/anticanres.16129
    OpenUrlAbstract/FREE Full Text
  17. ↵
    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
  18. ↵
    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
  19. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Hozumi C,
    6. Bouvet M,
    7. Yamamoto N,
    8. Hayashi K,
    9. Kimura H,
    10. Miwa S,
    11. Igarashi K,
    12. Higuchi T,
    13. Tsuchiya H,
    14. Demura S,
    15. Hoffman RM
    : Recombinant methioninase (rMETase) synergistically sensitizes ivermectin-resistant MCF-7 breast cancer cells 9.9 fold to low-dose ivermectin. Anticancer Res 45(2): 451-455, 2025. DOI: 10.21873/anticanres.17434
    OpenUrlAbstract/FREE Full Text
  20. ↵
    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
    : Ivermectin combined with recombinant methioninase (rMETase) synergistically eradicates MiaPaCa-2 pancreatic cancer cells. Anticancer Res 45(1): 97-103, 2025. DOI: 10.21873/anticanres.17396
    OpenUrlAbstract/FREE Full Text
  21. ↵
    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
  22. ↵
    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. 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): 429-434, 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
    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,
    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
    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. 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. 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
  23. ↵
    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
  24. ↵
    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
  25. ↵
    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
  26. ↵
    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
  27. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Yamamoto N,
    6. Hayashi K,
    7. Kimura H,
    8. Miwa S,
    9. Igarashi K,
    10. Higuchi T,
    11. Tsuchiya H,
    12. Demura S,
    13. Hoffman RM
    : Complete response (CR) in a previously-progressing chronic lymphocytic leukemia (CLL) patient treated with methionine restriction in combination with first-line chemotherapy. Cancer Diagn Progn 5(1): 21-26, 2025. DOI: 10.21873/cdp.10407
    OpenUrlCrossRefPubMed
    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
    : Synergistic eradication of fibrosarcoma with acquired ifosfamide resistance using methionine restriction combined with ifosfamide in nude-mouse models. In Vivo 39(1): 120-126, 2025. DOI: 10.21873/invivo.13809
    OpenUrlAbstract/FREE Full Text
  28. ↵
    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
  29. ↵
    1. Zhang P,
    2. Zhang Y,
    3. Liu K,
    4. Liu B,
    5. Xu W,
    6. Gao J,
    7. Ding L,
    8. Tao L
    : Ivermectin induces cell cycle arrest and apoptosis of HeLa cells via mitochondrial pathway. Cell Prolif 52(2): e12543, 2019. DOI: 10.1111/cpr.12543
    OpenUrlCrossRefPubMed
  30. ↵
    1. Hayashi A,
    2. Kamio K,
    3. Miyanaga A,
    4. Yoshida K,
    5. Noro R,
    6. Matsuda K,
    7. Tozuka T,
    8. Omori M,
    9. Hirao M,
    10. Fukuizumi A,
    11. Hisakane K,
    12. Takeuchi S,
    13. Matsumoto M,
    14. Kasahara K,
    15. Amano T,
    16. Honda K,
    17. Seike M
    : Ivermectin enhances paclitaxel efficacy by overcoming resistance through modulation of ABCB1 in non-small cell lung cancer. Anticancer Res 44(12): 5271-5282, 2024. DOI: 10.21873/anticanres.17355
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

Anticancer Research: 45 (6)
Anticancer Research
Vol. 45, Issue 6
June 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.
Selective Synergy of Ivermectin Combined With Recombinant Methioninase Against Colon-Cancer Cells in Contrast to Normal Fibroblasts
(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.
6 + 2 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Selective Synergy of Ivermectin Combined With Recombinant Methioninase Against Colon-Cancer Cells in Contrast to Normal Fibroblasts
YOHEI ASANO, QINGHONG HAN, SHUKUAN LI, KOHEI MIZUTA, BYUNG MO KANG, JIN SOO KIM, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, SEI MORINAGA, HIROYUKI TSUCHIYA, SATORU DEMURA, ROBERT M. HOFFMAN
Anticancer Research Jun 2025, 45 (6) 2257-2263; DOI: 10.21873/anticanres.17600

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Selective Synergy of Ivermectin Combined With Recombinant Methioninase Against Colon-Cancer Cells in Contrast to Normal Fibroblasts
YOHEI ASANO, QINGHONG HAN, SHUKUAN LI, KOHEI MIZUTA, BYUNG MO KANG, JIN SOO KIM, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, SEI MORINAGA, HIROYUKI TSUCHIYA, SATORU DEMURA, ROBERT M. HOFFMAN
Anticancer Research Jun 2025, 45 (6) 2257-2263; DOI: 10.21873/anticanres.17600
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

  • Tissue Prognostic Markers for Clear Cell Renal Cell Carcinoma Tumor-stroma Interaction: Impact on TNM Staging Parameters
  • CA9-Positive Keloid Fibroblasts: Modulator of Local Inflammation Induced by the Hypoxic and Glycolysis-enhanced Microenvironment
  • Predictive Factors for Multiple Metastases of Clear-cell Renal Cell Carcinoma
Show more Experimental Studies

Similar Articles

Keywords

  • Ivermectin
  • recombinant methioninase (rMETase)
  • combination
  • synergy
  • colorectal cancer
  • methionine addiction
  • Hoffman effect
  • normal fibroblasts
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire