Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues
  • 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
  • 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

Ivermectin Combined With Recombinant Methioninase (rMETase) Synergistically Eradicates MiaPaCa-2 Pancreatic Cancer Cells

SEI MORINAGA, QINGHONG HAN, KOHEI MIZUTA, BYUNG MO KANG, MICHAEL BOUVET, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, HIROYUKI TSUCHIYA, SATORU DEMURA and ROBERT M. HOFFMAN
Anticancer Research January 2025, 45 (1) 97-103; DOI: https://doi.org/10.21873/anticanres.17396
SEI MORINAGA
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
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
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
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
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{at}anticancer.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Ivermectin was initially utilized as a veterinary medication, demonstrating efficacy against various parasites. Pancreatic cancer is currently one of the most recalcitrant diseases. The aim of the present study was to demonstrate the synergy of the combination of recombinant methioninase (rMETase) and ivermectin to eradicate human pancreatic cancer cells in vitro. Materials and Methods: MiaPaCa-2 human pancreatic cancer cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) with the addition of 10% fetal bovine serum and 1 IU/ml penicillin/streptomycin. Reduction of cell viability by rMETase alone and ivermectin alone and their combination on MiaPaCa-2 cells was determined with the WST-reagent. Four experimental groups were examined in vitro: control group without treatment; ivermectin alone; rMETase alone; ivermectin combined with rMETase. Results: The IC50 of ivermectin for MiaPaCa-2 cells was 5.9 μM. The IC50 of rMETase on MiaPaCa-2 cells was 2.93 U/ml. Ivermectin (5.9 μM) plus rMETase (2.93 U/ml) synergistically greatly reduced the viability of MiaPaCa-2 cells, compared to ivermectin alone (80% reduction vs. 45% reduction, respectively p<0.05). Conclusion: The combination of ivermectin and rMETase effectively eradicated MiaPaCa-2 pancreatic cancer cells. The present results indicate the future clinical potential of the combination of rMETase, currently administered orally to patients as a dietary supplement, and oral ivermectin on pancreatic cancer.

Key Words:
  • Ivermectin
  • recombinant methioninase
  • synergy
  • pancreatic cancer
  • methionine addiction
  • Hoffman effect

Ivermectin was initially utilized as a veterinary medication and proved highly effective against various parasites, including gastrointestinal roundworms, lungworms, and mites (1). The Nobel prize was awarded to Satoshi Omura and William C. Campbell in 2015 for ivermectin as a novel therapy against roundworm parasites (1). Ivermectin induces cell-cycle arrest and apoptosis in HeLa cells through a mitochondrial pathway (2). Ivermectin also had anticancer efficacy against colon cancer, breast cancer, ovarian cancer, melanoma, and leukemia in pre-clinical studies (3).

Methionine addiction in cancer, also called the Hoffman Effect, is a fundamental and general hallmark of cancer (4). Numerous studies have shown that recombinant methioninase (rMETase), methionine-free medium, or a low-methionine diet are synergistic with chemotherapy for all major types of cancer, both in preclinical studies and in the clinic (5-24). Recently we have shown that the combination of rMETase with chemotherapy drugs can synergistically reverse cancer-cell resistance to drugs including eribulin, trabectedin, docetaxel, and doxorubicin (5, 12-13, 15, 16).

Pancreatic cancer is one of the most challenging cancers worldwide, highly aggressive and with poor prognosis. Targeted therapies, such as KRAS inhibitors have been evaluated in preclinical models and early-phase clinical trials for pancreatic cancer (25). Immune checkpoint inhibitors and CAR T-cell therapies, have been tested on pancreatic cancer cells (26). However, pancreatic cancer remains recalcitrant, with a high mortality rate and limited options, particularly for advanced stages (27). A recent clinical trial in metastatic pancreatic cancer showed that FOLFOX-6 (oxaliplatinum, leucovorin and 5-fluorouracil), added to standard therapy of nab-paclitaxel plus gemcitabine, increased median overall survival to 13.2 months compared to 9.7 months with standard therapy. However, treatment toxicity was greater with the new regimen (28). Therefore, there is a critical need for the development of new therapeutic strategies that offer improved efficacy and safety, as the majority of current treatments are associated with significant adverse effects and limited long-term outcomes.

The aim of the present study was to demonstrate the synergy of the combination of rMETase and ivermectin on human pancreatic-cancer cells, in vitro, as a new paradigm for pancreatic-cancer treatment.

Materials and Methods

Cell culture. MiaPaCa-2 pancreatic-cancer cells were obtained from the American Type Culture Collection (ATCC) (Manassas, VA, USA). MiaPaCa-2 cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) with the addition of 10% fetal bovine serum (FBS) and 1 IU/ml penicillin/streptomycin (10-013-CV; Corning, Corning, NY, USA).

Reagents. Ivermectin was obtained from MedChemExpress. (Monmouth Junction, NJ, USA). Recombinant methioninase (rMETase) was produced by AntiCancer Inc. as previously described (29).

Drug sensitivity assay 1: Determination of the IC50 of ivermectin and rMETase on MiaPaCa-2 pancreatic-cancer cells in vitro. Cell viability was determined with the WST-8 reagent (Dojindo Laboratory, Kumamoto, Japan). MiaPaCa-2 cells were grown in 96-well dishes, with 3×103 cells per well, in DMEM (100 μl per well). rMETase (0.5 U/ml to 8 U/ml) or ivermectin (5 μM to 80 μM) were added to the cells for 72 h. At the end of the culture period, 10 μl of the WST-8 solution was added to each well. Subsequently, the dish was incubated for 1 h at 37°C. Absorption was measured at 450 nm with a microplate reader (SUNRISE: TECAN, Mannedorf, Switzerland) which indicates the extent of viable cells. Microsoft Excel for Mac 2016 version 15.52 (Microsoft, Redmond, WA, USA) was used to generate drug-sensitivity curves. ImageJ version 1.53k (National Institutes of Health, Bethesda, MD, USA) was used to determine half-maximal inhibitory-concentrations (IC50) values. Each experiment was conducted twice in triplicate.

Drug sensitivity assay 2: Determination of synergy of ivermectin and rMETase on MiaPaCa-2 pancreatic cancer cells. MiaPaCa-2 cells were seeded in 96-well plates at a density of 3×103 cells per well. The cells were treated as follows 24 h later: Control (DMEM); ivermectin alone (5.9 μM [IC50]); rMETase alone (2.93 U/ml [IC50]); ivermectin (5.9 μM [IC50]) plus rMETase (2.93 U/ml [IC50]). The viability of the cells was evaluated 72 h later, as described above.

Statistical analysis. EZR software (Jichi Medical University, Saitama, Japan) was used for statistical analyses (30). A Tukey–Kramer analysis was used to analyze the relationship between variables and p-values ≤0.05 were considered statistically significant.

Results

IC50 of ivermectin alone and rMETase alone on MiaPaCa-2 pancreatic cancer cells. The IC50 value of ivermectin alone on MiaPaCa-2 cells was 5.9 μM. The IC50 for rMETase alone on MiaPaCa-2 cells was 2.93 U/ml (Figure 1).

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

IC50 determination of ivermectin and recombinant methioninase (rMETase) on MiaPaCa-2 pancreatic cancer cells. (A) IC50 of ivermectin on MiaPaCa-2 cells. (B) IC50 of rMETase on MiaPaCa-2 cells.

Synergy of rMETase plus ivermectin on MiaPaCa-2 pancreatic cancer cells. Compared to the control cells, treatment with ivermectin alone reduced the viability of MiaPaCa-2 cells by approximately 54%, rMETase alone reduced the viability of MiaPaCa-2 cells by 37%, and the combination of ivermectin (5.9 μM [IC50]) plus rMETase (2.93 U/ml [IC50]) caused a significantly greater reduction in the viability of MiaPaCa-2 cells, approximately 80% compared to ivermectin alone or rMETase alone, p<0.05 (Figure 2).

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

Efficacy of the combination of ivermectin and recombinant methioninase (rMETase) on MiaPaCa-2 pancreatic cancer cells. Control (DMEM); ivermectin (5.9 μM [IC50 for MiaPaCa-2 cells]); rMETase (2.93 U/ml [IC50 for MiaPaCa-2 cells]); ivermectin (5.9 μM [IC50 for MiaPaCa-2 cells]) plus rMETase (2.93 U/ml [IC50 for MiaPaCa-2 cells]).

Discussion

Pancreatic cancer ranks as the fourth most prevalent cause of cancer-related mortality globally and is one of the most recalcitrant cancers. The 5-year survival rate is approximately 10% due to late diagnosis, recurrent metastases, and restricted treatment alternatives (31-34). Consequently, it is essential to identify new chemotherapeutic agents and design effective therapeutic strategies.

Ivermectin is an antiparasitic medication approved by the U.S. Food and Drug Administration (FDA) for human use and is extensively utilized in animals (35). Ivermectin has shown efficacy against breast cancer, gastrointestinal cancer, urological cancer, hematological malignancies, reproductive-system cancer, glioma, pulmonary cancer, and melanoma as well as HeLa cells, in laboratory studies (36). Ivermectin reverses multidrug resistance, suppresses angiogenesis, and reduces mitochondrial biogenesis (37).

Methionine restriction (MR) by rMETase causes cancer cells to enter a reversible arrest in the late-S/G2-phase of the cell cycle where DNA damage occurs (38). Ivermectin induces G1/S phase cycle arrest in cancer cells, which can also cause DNA damage (2, 39). In the present study, the combination of rMETase and ivermectin showed synergistic efficacy against MiaPaCa-2 pancreatic cancer cells.

The main limitation of the current study is that it was performed in vitro, using a single cell line. While this cell line is commonly used in pancreatic-cancer research, relying on a single model limits the generalizability of our findings, as results may among different cancer cell lines. Furthermore, the molecular mechanisms underlying the observed synergistic efficacy were not explored in the present study, leaving unanswered questions regarding the specific pathways and interactions responsible for the treatment’s efficacy. Future studies should incorporate a broader range of cell lines, investigate the molecular mechanisms involved, and include toxicity assessments to provide a more comprehensive evaluation of the therapeutic potential and safety of the present approach.

However the present results demonstrate the therapeutic potential of ivermectin plus rMETase, as combination treatment for pancreatic-cancer patients.

rMETase is effective because it targets the fundamental hallmark of cancer, methionine addiction (4, 38, 40-66).

Acknowledgements

This article is dedicated to the memory of A.R. Moossa, MD, Sun Lee, MD, Professor Gordon H. Sato, Professor Li Jiaxi, Masaki Kitajima, MD, Joseph R. Bertino, MD, Shigeo Yagi, PhD, J.A.R Mead, Ph.D., Eugene P. Frenkel, MD, Professor Lev Bergelson, Professor Sheldon Penman, Professor John R. Raper, Joseph Leighton, MD and John Mendelsohn, MD.

Footnotes

  • Authors’ Contributions

    SM, RMH, and QH designed the study. QH provided rMETase. SM performed experiments. SM was the major contributor to writing the article and RMH revised the article. KM, BMK, MB, NY, KH, HK, SM, KI, TH, HT, QH, and SD critically read the manuscript.

  • Conflicts of Interest

    The Authors have declared that there are no competing interests in relation to this study.

  • Received October 13, 2024.
  • Revision received November 10, 2024.
  • Accepted November 15, 2024.
  • 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. Campbell WC
    : Ivermectin: a reflection on simplicity (Nobel lecture). Angew Chem Int Ed Engl 55(35): 10184-10189, 2016. DOI: 10.1002/anie.201601492
    OpenUrlCrossRef
  2. ↵
    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
  3. ↵
    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
  4. ↵
    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
  5. ↵
    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
    : Selective Synergy of recombinant methioninase plus docetaxel against docetaxel-resistant and -sensitive fibrosarcoma cells compared to normal fibroblasts. Anticancer Res 44(12): 5207-5213, 2024. DOI: 10.21873/anticanres.17347
    OpenUrlAbstract/FREE Full Text
    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
    1. Morinaga S,
    2. Han Q,
    3. Kubota Y,
    4. Mizuta K,
    5. Kang BM,
    6. Sato M,
    7. Bouvet M,
    8. Yamamoto N,
    9. Hayashi K,
    10. Kimura H,
    11. Miwa S,
    12. Igarashi K,
    13. Higuchi T,
    14. Tsuchiya H,
    15. Hoffman RM
    : Extensive synergy between recombinant methioninase and eribulin against fibrosarcoma cells but not normal fibroblasts. Anticancer Res 44(3): 921-928, 2024. DOI: 10.21873/anticanres.16886
    OpenUrlAbstract/FREE Full Text
    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
    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. 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
    1. Kubota Y,
    2. Aoki Y,
    3. Masaki N,
    4. Obara K,
    5. Hamada K,
    6. Han Q,
    7. Bouvet M,
    8. Tsunoda T,
    9. Hoffman RM
    : Methionine restriction of glioma does not induce MGMT and greatly improves temozolomide efficacy in an orthotopic nude-mouse model: A potential curable approach to a clinically-incurable disease. Biochem Biophys Res Commun 695: 149418, 2024. DOI: 10.1016/j.bbrc.2023.149418
    OpenUrlCrossRefPubMed
  6. ↵
    1. Morinaga S,
    2. Han Q,
    3. Kubota Y,
    4. Mizuta K,
    5. Kang BM,
    6. Sato M,
    7. Bouvet M,
    8. Yamamoto N,
    9. Hayashi K,
    10. Kimura H,
    11. Miwa S,
    12. Igarashi K,
    13. Higuchi T,
    14. Tsuchiya H,
    15. Demura S,
    16. Hoffman RM
    : 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. Anticancer Res 44(6): 2359-2367, 2024. DOI: 10.21873/anticanres.17043
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Sato M,
    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 is selectively synergistic with doxorubicin against wild-type fibrosarcoma cells compared to normal cells and overcomes highly-doxorubicin-resistant fibrosarcoma. Anticancer Res 44(8): 3261-3268, 2024. DOI: 10.21873/anticanres.17144
    OpenUrlAbstract/FREE Full Text
    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
  8. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Sato M,
    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
    : Overcoming high trabectedin resistance of soft-tissue sarcoma with recombinant methioninase: a potential solution of a recalcitrant clinical problem. Anticancer Res 44(9): 3785-3791, 2024. DOI: 10.21873/anticanres.17203
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Sato M,
    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 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. Anticancer Res 44(9): 3777-3783, 2024. DOI: 10.21873/anticanres.17202
    OpenUrlAbstract/FREE Full Text
    1. Sato M,
    2. Han Q,
    3. Mori R,
    4. Mizuta K,
    5. Kang BM,
    6. Morinaga S,
    7. Kobayashi N,
    8. Ichikawa Y,
    9. Nakajima A,
    10. Hoffman RM
    : 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. Anticancer Res 44(4): 1499-1504, 2024. DOI: 10.21873/anticanres.16946
    OpenUrlAbstract/FREE Full Text
    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
    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. 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. 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. 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
    1. Sato M,
    2. Han Q,
    3. Mizuta K,
    4. Mori R,
    5. Kang BM,
    6. Morinaga S,
    7. Kobayashi N,
    8. Ichikawa Y,
    9. Nakajima A,
    10. Hoffman RM
    : Extensive shrinkage and long-term stable disease in a teenage female patient with high-grade glioma treated with temozolomide and radiation in combination with oral recombinant methioninase and a low-methionine diet. In Vivo 38(3): 1459-1464, 2024. DOI: 10.21873/invivo.13591
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Sato M,
    2. Han Q,
    3. Hozumi C,
    4. Kujiraoka H,
    5. Mizuta K,
    6. Morinaga S,
    7. Kang BM,
    8. Kobayashi N,
    9. Ichikawa Y,
    10. Nakajima A,
    11. Hoffman RM
    : First-line chemotherapy in combination with oral recombinant methioninase and a low-methionine diet for a stage IV inoperable pancreatic-cancer patient resulted in 40% tumor reduction and an 86% CA19-9 biomarker decrease. Anticancer Res 44(9): 3885-3889, 2024. DOI: 10.21873/anticanres.17215
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Linehan A,
    2. O’Reilly M,
    3. McDermott R,
    4. O’Kane GM
    : Targeting KRAS mutations in pancreatic cancer: opportunities for future strategies. Front Med (Lausanne) 11: 1369136, 2024. DOI: 10.3389/fmed.2024.1369136
    OpenUrlCrossRefPubMed
  12. ↵
    1. Cutmore LC,
    2. Brown NF,
    3. Raj D,
    4. Chauduri S,
    5. Wang P,
    6. Maher J,
    7. Wang Y,
    8. Lemoine NR,
    9. Marshall JF
    : Pancreatic Cancer UK Grand Challenge: Developments and challenges for effective CAR T cell therapy for pancreatic ductal adenocarcinoma. Pancreatology 20(3): 394-408, 2020. DOI: 10.1016/j.pan.2020.02.006
    OpenUrlCrossRefPubMed
  13. ↵
    1. Siegel RL,
    2. Miller KD,
    3. Wagle NS,
    4. Jemal A
    : Cancer statistics, 2023. CA Cancer J Clin 73(1): 17-48, 2023. DOI: 10.3322/caac.21763
    OpenUrlCrossRefPubMed
  14. ↵
    1. Carrato A,
    2. Pazo-Cid R,
    3. Macarulla T,
    4. Gallego J,
    5. Jiménez-Fonseca P,
    6. Rivera F,
    7. Cano MT,
    8. Rodriguez-Garrote M,
    9. Pericay C,
    10. Alés I,
    11. Layos L,
    12. Graña B,
    13. Iranzo V,
    14. Gallego I,
    15. Garcia-Carbonero R,
    16. de Mena IR,
    17. Guillén-Ponce C,
    18. Aranda E
    : Nab-paclitaxel plus gemcitabine and FOLFOX in metastatic pancreatic cancer. NEJM Evid 3(2): EVIDoa2300144, 2024. DOI: 10.1056/EVIDoa2300144
    OpenUrlCrossRef
  15. ↵
    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 recombinant l-methionine-alpha-deamino-gamma-mercaptomethane-lyase for novel anticancer therapy. Protein Expr Purif 9(2): 233-245, 1997. DOI: 10.1006/prep.1996.0700
    OpenUrlCrossRefPubMed
  16. ↵
    1. Kanda Y
    : Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant 48(3): 452-458, 2013. DOI: 10.1038/bmt.2012.244
    OpenUrlCrossRefPubMed
  17. ↵
    1. Jia Y,
    2. Gu D,
    3. Wan J,
    4. Yu B,
    5. Zhang X,
    6. Chiorean EG,
    7. Wang Y,
    8. Xie J
    : The role of GLI-SOX2 signaling axis for gemcitabine resistance in pancreatic cancer. Oncogene 38(10): 1764-1777, 2019. DOI: 10.1038/s41388-018-0553-0
    OpenUrlCrossRefPubMed
    1. Hyatt HW,
    2. Powers SK
    : Mitochondrial dysfunction is a common denominator linking skeletal muscle wasting due to disease, aging, and prolonged inactivity. Antioxidants (Basel) 10(4): 588, 2021. DOI: 10.3390/antiox10040588
    OpenUrlCrossRef
    1. Lee KH,
    2. Chie EK,
    3. Im SA,
    4. Kim JH,
    5. Kwon J,
    6. Han SW,
    7. Oh DY,
    8. Jang JY,
    9. Kim JS,
    10. Kim TY,
    11. Bang YJ,
    12. Kim SW,
    13. Ha SW
    : Phase II trial of postoperative adjuvant gemcitabine and cisplatin chemotherapy followed by chemoradiotherapy with gemcitabine in patients with resected pancreatic cancer. Cancer Res Treat 53(4): 1096-1103, 2021. DOI: 10.4143/crt.2020.928
    OpenUrlCrossRefPubMed
  18. ↵
    1. Liang C,
    2. Shi S,
    3. Meng Q,
    4. Liang D,
    5. Ji S,
    6. Zhang B,
    7. Qin Y,
    8. Xu J,
    9. Ni Q,
    10. Yu X
    : Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going. Exp Mol Med 49(12): e406, 2017. DOI: 10.1038/emm.2017.255
    OpenUrlCrossRefPubMed
  19. ↵
    1. Zhang J,
    2. Yan YJ,
    3. An J,
    4. Huang SX,
    5. Wang XJ,
    6. Xiang WS
    : Designed biosynthesis of 25-methyl and 25-ethyl ivermectin with enhanced insecticidal activity by domain swap of avermectin polyketide synthase. Microb Cell Fact 14: 152, 2015. DOI: 10.1186/s12934-015-0337-y
    OpenUrlCrossRefPubMed
  20. ↵
    1. Tang M,
    2. Hu X,
    3. Wang Y,
    4. Yao X,
    5. Zhang W,
    6. Yu C,
    7. Cheng F,
    8. Li J,
    9. Fang Q
    : Ivermectin, a potential anticancer drug derived from an antiparasitic drug. Pharmacol Res 163: 105207, 2021. DOI: 10.1016/j.phrs.2020.105207
    OpenUrlCrossRefPubMed
  21. ↵
    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
  22. ↵
    1. Hoffman RM,
    2. Jacobsen SJ
    : Reversible growth arrest in simian virus 40-transformed human fibroblasts. Proc Natl Acad Sci U S A 77(12): 7306-7310, 1980. DOI: 10.1073/pnas.77.12.7306
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Zhou S,
    2. Wu H,
    3. Ning W,
    4. Wu X,
    5. Xu X,
    6. Ma Y,
    7. Li X,
    8. Hu J,
    9. Wang C,
    10. Wang J
    : Ivermectin has new application in inhibiting colorectal cancer cell growth. Front Pharmacol 12: 717529, 2021. DOI: 10.3389/fphar.2021.717529
    OpenUrlCrossRefPubMed
  24. ↵
    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. Kaiser P
    : Methionine dependence of cancer. Biomolecules 10(4): 568, 2020. DOI: 10.3390/biom10040568
    OpenUrlCrossRefPubMed
    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
    : 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. 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. 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
    : Deletion of MTAP highly sensitizes osteosarcoma cells to methionine restriction with recombinant methioninase. Cancer Genomics Proteomics 19(3): 299-304, 2022. DOI: 10.21873/cgp.20321
    OpenUrlAbstract/FREE Full Text
    1. Aoki Y,
    2. Tome Y,
    3. Han Q,
    4. Yamamoto J,
    5. Hamada K,
    6. Masaki N,
    7. Bouvet M,
    8. Nishida K,
    9. Hoffman RM
    : Histone H3 lysine-trimethylation markers are decreased by recombinant methioninase and increased by methotrexate at concentrations which inhibit methionine-addicted osteosarcoma cell proliferation. Biochem Biophys Rep 28: 101177, 2021. DOI: 10.1016/j.bbrep.2021.101177
    OpenUrlCrossRefPubMed
    1. Aoki Y,
    2. Yamamoto J,
    3. Tome Y,
    4. Hamada K,
    5. Masaki N,
    6. Inubushi S,
    7. Tashiro Y,
    8. Bouvet M,
    9. Endo I,
    10. Nishida K,
    11. Hoffman RM
    : Over-methylation of histone H3 lysines is a common molecular change among the three major types of soft-tissue sarcoma in patient-derived xenograft (PDX) mouse models. Cancer Genomics Proteomics 18(6): 715-721, 2021. DOI: 10.21873/cgp.20292
    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 76(3): 1313-1317, 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. 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: 1034-1038, 2020. DOI:10.1016/j.bbrc.2020.09.108
    OpenUrlCrossRefPubMed
    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
    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
    1. Hoffman RM
    : Altered methionine metabolism, DNA methylation and oncogene expression in carcinogenesis. A review and synthesis. Biochim Biophys Acta 738: 49-87, 1984. DOI: 10.1016/0304-419x(84)90019-2
    OpenUrlCrossRefPubMed
    1. Ghergurovich JM,
    2. Xu X,
    3. Wang JZ,
    4. Yang L,
    5. Ryseck RP,
    6. Wang L,
    7. Rabinowitz JD
    : Methionine synthase supports tumour tetrahydrofolate pools. Nat Metab 3(11): 1512-1520, 2021. DOI: 10.1038/s42255-021-00465-w
    OpenUrlCrossRefPubMed
    1. Sullivan MR,
    2. Darnell AM,
    3. Reilly MF,
    4. Kunchok T,
    5. Joesch-Cohen L,
    6. Rosenberg D,
    7. Ali A,
    8. Rees MG,
    9. Roth JA,
    10. Lewis CA,
    11. Vander Heiden MG
    : Methionine synthase is essential for cancer cell proliferation in physiological folate environments. Nat Metab 3(11): 1500-1511, 2021. DOI: 10.1038/s42255-021-00486-5
    OpenUrlCrossRef
    1. Mecham JO,
    2. Rowitch D,
    3. Wallace C,
    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. 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
    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
    1. Hoffman RM,
    2. Coalson DW,
    3. Jacobsen SJ,
    4. Erbe RW
    : Folate polyglutamate and monoglutamate accumulation in normal and SV40-transformed human fibroblasts. J Cell Physiol 109(3): 497-505, 1981. DOI: 10.1002/jcp.1041090316
    OpenUrlCrossRefPubMed
    1. Oden KL,
    2. Carson K,
    3. Mecham JO,
    4. Hoffman RM,
    5. Clarke S
    : S-adenosylmethionine synthetase in cultured normal and oncogenically-transformed human and rat cells. Biochim Biophys Acta 760(2): 270-277, 2983. DOI: 10.1016/0304-4165(83)90173-3
    OpenUrlCrossRef
    1. Rubnitz JE,
    2. Jacobsen SJ,
    3. Hoffman RM
    : Constitutive behavior of methionyl-tRNA synthetase compared to repressible behavior of methionine adenosyltransferase in mammalian cells. Biochim Biophys Acta 677(2): 269-273, 1981. DOI: 10.1016/0304-4165(81)90095-7
    OpenUrlCrossRefPubMed
    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: 10.1517/14712598.20
    OpenUrlCrossRefPubMed
  25. ↵
    1. Jacobsen SJ,
    2. Hoffman RM,
    3. Erbe RW
    : Regulation of methionine adenosyltransferase in normal diploid and simian virus 40-transformed human fibroblasts. J Natl Cancer Inst 65(6): 1237-1244, 1980. PMID: 6253712.
    OpenUrlPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 45 (1)
Anticancer Research
Vol. 45, Issue 1
January 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.
Ivermectin Combined With Recombinant Methioninase (rMETase) Synergistically Eradicates MiaPaCa-2 Pancreatic 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.
11 + 5 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Ivermectin Combined With Recombinant Methioninase (rMETase) Synergistically Eradicates MiaPaCa-2 Pancreatic Cancer Cells
SEI MORINAGA, QINGHONG HAN, KOHEI MIZUTA, BYUNG MO KANG, MICHAEL BOUVET, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, HIROYUKI TSUCHIYA, SATORU DEMURA, ROBERT M. HOFFMAN
Anticancer Research Jan 2025, 45 (1) 97-103; DOI: 10.21873/anticanres.17396

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Ivermectin Combined With Recombinant Methioninase (rMETase) Synergistically Eradicates MiaPaCa-2 Pancreatic Cancer Cells
SEI MORINAGA, QINGHONG HAN, KOHEI MIZUTA, BYUNG MO KANG, MICHAEL BOUVET, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, HIROYUKI TSUCHIYA, SATORU DEMURA, ROBERT M. HOFFMAN
Anticancer Research Jan 2025, 45 (1) 97-103; DOI: 10.21873/anticanres.17396
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

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

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Triple Combination of Recombinant Methioninase and the Anti-parasitic Drugs Ivermectin, and Chloroquine Selectively Eradicates Pancreatic Cancer Cells While Sparing Normal Fibroblasts
  • Selective Synergy of Ivermectin Combined With Recombinant Methioninase Against Colon-Cancer Cells in Contrast to Normal Fibroblasts
  • Recombinant Methioninase and Cisplatinum Act Synergistically to Inhibit Lewis Lung Carcinoma Cells But Not Normal Fibroblasts
  • Recombinant Methioninase (rMETase) Synergistically Sensitizes Ivermectin-resistant MCF-7 Breast Cancer Cells 9.9 Fold to Low-dose Ivermectin
  • Google Scholar

More in this TOC Section

  • Pulsed Electromagnetic Field Promotes Doxorubicin-induced Apoptosis by Increasing Caspase-2 Activation in MDA-MB-231 Breast Cancer Cells
  • Fibroblast Supernatants Modulate Treatment Responses in Human Papillomavirus Positive and Negative Oropharyngeal Cancer Cell Lines
  • Impact of Interleukin-12B Genotypes on Breast Cancer Risk
Show more Experimental Studies

Similar Articles

Keywords

  • ivermectin
  • recombinant methioninase
  • synergy
  • pancreatic cancer
  • methionine addiction
  • Hoffman effect
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire