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

Triple Combination of Recombinant Methioninase and the Anti-parasitic Drugs Ivermectin, and Chloroquine Selectively Eradicates Pancreatic Cancer Cells While Sparing Normal Fibroblasts

YOHEI ASANO, QINGHONG HAN, SHUKUAN LI, KOHEI MIZUTA, BYUNG MO KANG, JIN SOO KIM, YUTA MIYASHI, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, SEI MORINAGA, HIROYUKI TSUCHIYA, SATORU DEMURA and ROBERT M. HOFFMAN
Anticancer Research November 2025, 45 (11) 4791-4802; DOI: https://doi.org/10.21873/anticanres.17828
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
YUTA MIYASHI
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{at}anticancer.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Pancreatic cancer is a recalcitrant disease which often presents with few symptoms in the early stages and is frequently diagnosed with distant metastases, limiting treatment options and resulting in a poor prognosis. Recombinant methioninase (rMETase), which targets cancer-specific methionine addiction, has shown synergistic efficacy with numerous types of chemotherapy against all major cancer types. Ivermectin and chloroquine, anti-parasitic drugs, are showing promise against cancer. The present study investigates in vitro the potential synergy of rMETase combined with ivermectin and chloroquine, two agents with multiple anticancer mechanisms, as a novel treatment strategy for metastatic pancreatic cancer.

Materials and Methods: The human pancreatic-cancer cell line MiaPaCa-2 and normal human fibroblasts Hs27 were cultured in 96-well plates (1×103 cells/well) for 24 h. Cell viability was assessed using the WST-8 reagent following 72-h treatment with rMETase, ivermectin, or chloroquine to determine their 30% inhibitory concentration (IC30) values. To evaluate synergy, cells were treated with each drug alone or double or triple combinations at their respective IC30 concentrations. Additionally, to evaluate the optimal order of the combination therapy, MiaPaCa-2 cells were divided into four groups and sequentially treated for 72 h as follows: (1) untreated control; (2) the triple-drug combination therapy alone; (3) rMETase followed by the triple-drug combination therapy; (4) the triple-drug combination therapy followed by rMETase.

Results: The IC30 value of rMETase was 0.39 U/ml, ivermectin was 4.41 μM, and chloroquine was 3.29 μM on MiaPaCa-2 cells. The triple combination of these agents at their IC30 concentrations significantly inhibited MiaPaCa-2 cell growth compared to monotherapies or dual combinations, indicating a synergistic efficacy. In contrast, the same combination had minimal impact on Hs27 cells at the IC30 values determined for MiaPaCa-2. Regarding the treatment sequence, triple-drug combination therapy and triple-drug combination treatment followed by rMETase treatment significantly inhibited cell proliferation more than rMETase followed by the triple-drug combination treatment.

Conclusion: The combination of rMETase, ivermectin, and chloroquine exhibited a selective cytotoxic synergy against pancreatic-cancer cells while sparing normal fibroblasts. Furthermore, the order of treatment may also affect efficacy. This triple combination treatment may offer a novel and effective first-line therapeutic approach for pancreatic cancer.

Keywords:
  • Recombinant methioninase (rMETase)
  • ivermectin
  • chloroquine
  • combination treatment
  • synergy
  • pancreatic cancer
  • normal fibroblasts
  • methionine addiction
  • Hoffman effect

Introduction

Pancreatic cancer is a recalcitrant disease and the 7th leading cause of cancer-related deaths worldwide (1). The estimated 5-year survival rate is approximately 10%, which has not significantly changed in decades (1). The poor prognosis is largely attributable to the asymptomatic nature of early-stage disease, leading to delayed diagnosis and a high incidence of recalcitrant distant metastases at initial presentation (2).

Targeting methionine addiction, a fundamental and general hallmark of cancer known as the Hoffman effect, has shown preclinical efficacy and clinical promise (3-14). Previous studies have shown potent efficacy of recombinant methioninase (rMETase) against all major cancer types, including pancreatic cancer, in both in vitro and in vivo models and the clinic (7, 10). Furthermore, rMETase has shown potential for synergy when combined with numerous types of chemotherapy in preclinical studies and clinical case reports of major cancer types (7, 10-14).

Recently, we have combined rMETase with ivermectin, which is conventionally used as an anti-parasitic agent in clinical practice (15-19). Ivermectin also has anti-cancer efficacy through multiple mechanisms, including induction of apoptosis, modulating autophagy, and inhibiting tumor proliferation and metastasis (15, 20-25). A recent in vitro study showed that the combination of rMETase with ivermectin eradicated pancreatic cancer cells (26).

Chloroquine, an anti-malarial drug, has shown efficacy on several malignancies through autophagy inhibition and other mechanisms (27-31). Previous studies have reported the efficacy of chloroquine on pancreatic cancer (32-36). We have previously demonstrated that the combination of rMETase with chloroquine acted synergistically on colorectal cancer cells (37, 38), but its efficacy on pancreatic cancer is unknown.

In the present study, we aimed to determine the synergistic efficacy of a triple-drug combination of rMETase, ivermectin, and chloroquine against a pancreatic-cancer cell line compared to normal fibroblasts.

Materials and Methods

Cell culture. The human pancreatic-cancer cell line MiaPaCa-2 and normal human fibroblasts Hs27 were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). Cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin–streptomycin, and maintained at 37°C in a humidified atmosphere containing 5% CO2.

Recombinant methioninase (rMETase) and drugs. rMTEase was produced as previously reported (39) at AntiCancer Inc (San Diego, CA, USA). Ivermectin and chloroquine diphosphate were obtained from MedChemoExpress (Monmouth Junction, NJ, USA) and Sigma-Aldrich (St. Louis, MO, USA), respectively.

Cell viability assay and 30% inhibitory concentration (IC30) determination. MiaPaCa-2 and Hs27 cells were seeded into 96-well plates at a density of 1×103 cells per well in 100 μl DMEM and cultured for 24 h. After confirming cell adhesion and proliferation, the cells were treated with rMETase at concentrations ranging from 0.0625 to 8 U/ml, and with either ivermectin or chloroquine at concentrations ranging from 1 to 128 μM for 72 h. Subsequently, to measure cell viability; the WST-8 reagent (10 μl) (Dojindo Laboratories, Kumamoto, Japan) was added to each well, followed by incubation for 1 h. Absorbance was then measured at 450 nm using a microplate reader (Sunrise, Tecan, Männedorf, Switzerland).

Based on the absorbance data, drug-sensitivity curves for rMETase, ivermectin, and chloroquine were generated for both MiaPaCa-2 and Hs27 cells using Microsoft Excel for Mac 2024 (ver. 16.89.1; Microsoft, Redmond, WA, USA) and ImageJ (ver. 1.54g; National Institutes of Health, Bethesda, MD, USA). The 30% inhibitory-concentration (IC30) values were subsequently calculated from these curves. In addition, the half-maximal inhibitory concentration (IC50) of rMETase was also determined for use in treatment-sequence experiments.

Evaluation of synergy between rMETase, ivermectin, and chloroquine on MiaPaCa-2 and Hs27 cells. MiaPaCa-2 and Hs27 cells were seeded into 96-well plates and cultured for 24 h following the procedure described above. Subsequently, the cells were divided into eight groups and treated for 72 h. Each treatment of MiaPaCa-2 cells and Hs27 fibroblasts was performed using the IC30 value previously determined for each agent on MiaPaCa-2 cells. The experimental groups were as follows: 1) control (DMEM), 2) rMETase alone (IC30), 3) ivermectin alone (IC30), 4) chloroquine alone (IC30), 5) rMETase + ivermectin (each at IC30), 6) rMETase + chloroquine (each at IC30), 7) ivermectin + chloroquine (each at IC30), 8) rMETase + ivermectin + chloroquine (each at IC30). In addition, Hs27 fibroblasts were also treated with each agent at the IC30 values for Hs27 fibroblasts. After treatment, absorbance was measured, and the relative cell viability of each treatment group was calculated compared to the control.

Determination of optimal order of the triple-combination and rMETase alone. MiaPaCa-2 cells were seeded and cultured in 96-well plates following the procedure as described above. The cells were then divided into four groups and treated for 72 h each, as follows: 1) control (DMEM), 2) triple-drug combination (each at IC30), 3) rMETase (IC50) followed by the triple-drug combination (each at IC30), 4) the triple-drug combination (each at IC30) followed by rMETase (IC50). At the time of medium exchange after the first 72-h treatment, the cells were washed with phosphate-buffered saline (PBS). After 144 hours of total treatment, the WST-8 reagent was added, absorbance was measured as described above, and the relative cell viability was calculated for each treatment group compared to the control group.

Statistical analysis. All experiments in the present study were performed in triplicate and independently repeated twice. All data are presented as the mean±standard deviation (SD). Statistical comparisons between the treatment groups were conducted using one-way analysis of variance (ANOVA), followed by Tukey’s post hoc test for multiple comparisons. All statistical analyses were performed using EZR (Saitama Medical Center, Jichi Medical University, Saitama, Japan), a graphical user interface for R. A p-value of ≤0.05 was considered statistically significant.

Results

IC30 of rMETase, ivermectin, and chloroquine οn MiaPaCa-2 pancreatic-cancer cells and Hs27 normal fibroblasts. Drug-sensitivity curves of rMEase, ivermectin, and chloroquine for MiaPaCa-2 and Hs27 cells were generated based on cell-viability assays and their IC30 values were determined (Figure 1). The IC30 of rMETase was 0.39 U/ml, ivermectin 4.41 μM, and chloroquine 3.29 μM οn MiaPaCa-2 cells. The IC30 of rMETase was 0.50 U/ml, ivermectin was 4.62 μM, and chloroquine was 3.58 μM οn Hs27 fibroblasts. In addition, the IC50 of rMETase on MiaPaCa-2 cells was 0.58 U/ml (Table I).

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

Drug-sensitivity curves and 30% inhibitory concentration (IC30) of recombinant methioninase (rMETase), ivermectin (IVM), and chloroquine (CQ) on MiaPaCa-2 and Hs27 cells.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

The 30% inhibitory concentration (IC30) values of recombinant methioninase (rMETase), ivermectin (IVM), and chloroquine (CQ) on pancreatic cancer cells (MiaPaCa-2) and normal fibroblasts (Hs27).

Synergy of triple combination of rMETase, ivermectin, and chloroquine on MiaPaCa-2 cells and Hs27 fibroblasts. MiaPaCa-2 pancreatic cancer cells and Hs27 fibroblasts were treated with each drug at its specific IC30 concentration, either as monotherapy or in combination. The triple combination of rMETase, ivermectin, and chloroquine demonstrated synergistic efficacy against MiaPaCa-2 cells compared to each monotherapy or dual combination using the MiaPaCa-2 IC30 values for each agent (Figure 2A). In contrast, this triple combination did not show significant synergistic efficacy on Hs27 cells using the Hs27 IC30 values for each agent (Figure 2B). Moreover, when Hs27 cells were treated using the IC30 values determined for MiaPaCa-2 cells, minimal cytotoxicity was observed (Figure 2C and Table II), further suggesting the selective efficacy of the combination on the cancer cells.

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

Treatment with ivermectin, chloroquine and recombinant methioninase on MiaPaCa-2 pancreatic cancer cells and normal Hs27 fibroblasts. (A) Viability of MiaPaCa-2 cells treated with recombinant methioninase (rMETase, at IC30 for MiaPaCa-2 cells), ivermectin (IVM, at IC30 for MiaPaCa-2 cells), chloroquine (CQ, at IC30 for MiaPaCa-2 cells), rMETase plus IVM (at each IC30 for MiaPaCa-2 cells), rMETase plus CQ (at each IC30 for MiaPaCa-2 cells), IVM plus CQ (at each IC30 for MiaPaCa-2 cells), and rMETase plus IVM plus CQ (at each IC30 for MiaPaCa-2 cells). (B) Viability of Hs27 cells treated as in (A) but using the IC30 value of each agent on Hs27 fibroblasts. (C) Viability of Hs27 cells treated as in (A), using the IC30 for each agent determined for MiaPaCa-2 cells. *p-Value <0.05, ns: Not significant.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Cell-proliferation inhibitory rate of recombinant methioninase (rMETase), ivermectin (IVM), chloroquine (CQ) on MiaPaCa-2 and Hs27 cells treated with each drug alone and in combination.

Optimal order of the triple combination therapy. Among the treatment groups (Figure 3), continuous administration of the triple combination from the beginning resulted in the greatest inhibition of MiaPaCa-2 cell viability. The group treated with the triple drug combination followed by rMETase showed the second-highest inhibitory effect. There was no significant difference between these two groups (p=0.26). Both of these groups demonstrated significantly greater cell-proliferation inhibition compared to the group treated with rMETase followed by the triple-drug combination therapy (Figure 4, both p<0.05).

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

Determination of the optimal order of combination therapy (Comb) with recombinant methioninase (rMETase) alone, ivermectin (IVM), and chloroquine (CQ) on MiaPaCa-2 cells. rMETase also was administered at its IC50 for MiaPaCa-2 cells, while the triple-drug combination of rMETase, ivermectin and chloroquine was applied using the IC30 of each drug against MiaPaCa-2 cells. Each treatment was carried out for 72 h, and cell viability was subsequently assessed as described in the Materials and Methods. IVM: Ivermectin; CQ: chloroquine.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Efficacy of treatment sequence of recombinant methioninase (rMETase) alone and the triple-drug combination (Comb) of rMETase, ivermectin and chloroquine on the viability of MiaPaCa-2 cells. Please see Figure 3 for treatment schema. *p-Value <0.05, ns: Not significant.

Discussion

The present results demonstrated that the triple combination of rMETase, ivermectin, and chloroquine exerts selective synergistic cytotoxic efficacy on pancreatic cancer cells (MiaPaCa-2) in contrast to normal fibroblasts.

rMETase targets methionine addiction, a fundamental and general hallmark of cancer (3-6). Its efficacy has been demonstrated in both in vitro and in vivo studies on all major cancer types, including pancreatic cancer (7, 10). rMETase selectively arrests the cancer cell cycle at the S/G2 phase (40, 41). Consequently, rMETase has been shown to act synergistically with chemotherapeutic agents targeting the S-phase of the cell cycle, suggesting its potential as additive therapy to enhance the efficacy of chemotherapy (3, 7, 10, 42-47), especially in malignancies with poor prognosis such as metastatic pancreatic cancer.

Ivermectin is a 16-membered macrocyclic lactone that has been used clinically as an anti-parasitic agent (15-19). However, recent studies have reported its anti-cancer properties through multiple mechanisms, including the induction of caspase-dependent apoptosis, inhibition of cance-cell proliferation via p21-activated kinase 1 (PAK1)-mediated autophagy, cell-cycle arrest in the S phase, and reversal of chemo-resistance (15, 20-25, 48). In vitro efficacy of ivermectin has been demonstrated in various cancers, including cholangiocarcinoma (22), lung cancer (49), breast cancer (21, 23), colorectal cancer, and leukemia (50, 51).

Similarly, chloroquine, originally developed as an anti-malarial drug, exerts diverse anti-cancer mechanisms. Previous studies have shown that chloroquine induces G1 cell-cycle arrest by inhibiting autophagy, promotes apoptosis via activation of the caspase cascade, and suppresses tumor invasion and metastasis by normalizing the structure and function of tumor vasculature (28, 30, 31, 52, 53) demonstrated in multiple malignancies such as primary effusion lymphoma (29), lung cancer (54), pancreatic cancer (32-36), colorectal cancer (37, 38, 52, 53, 55), breast cancer (56), and osteosarcoma (57). Especially, chloroquine has been used in combination with chemotherapy for pancreatic cancer both pre-clinically and clinically (32-36).

The combination of rMETase with ivermectin previously showed synergy on pancreatic, breast, and colorectal cancer cells in vitro (26, 51, 58), and the combination of rMETase with chloroquine showed synergy on breast cancer, osteosarcoma, and colorectal cancer cells in vitro (37, 56, 57). In the present study, we provide the first evidence that the triple combination of these agents exerts selective synergistic efficacy on pancreatic-cancer cells, in contrast to normal fibroblasts.

Both rMETase and ivermectin target the S-phase of the cell cycle, suggesting the potential for enhanced S-phase inhibition when used in combination. Moreover, since chloroquine targets the G1-phase, the triple combination may exert broad cell cycle inhibition, which could contribute to the observed synergy. Both ivermectin and chloroquine are known to induce apoptosis through modulation of autophagy (15, 21, 23, 27, 29, 32). Methionine restriction itself has also been suggested to influence autophagy (59, 60), and the impact of this triple combination on autophagic pathways may be another possible mechanism underlying the synergy.

Although the synergy of rMETase in combination with chemotherapeutic agents has been demonstrated in various studies, the optimal order of such combination treatments remains largely unexplored. In a previous study investigating the sequential administration of rMETase and rapamycin in colorectal cancer HCT-116 cells, the cells that received continuous combination treatment had the greatest inhibition of proliferation. The group treated with rapamycin followed by rMETase showed the second highest inhibitory effect (61), consistent with the present findings. While the underlying mechanisms of treatment sequence–dependent efficacy were not elucidated in the present study, our results suggest that initiating triple combination therapy as a first-line treatment may be effective in the clinical setting. Additionally, in cases where the combination therapy must be discontinued due to adverse effects, continuing rMETase mono-therapy may still have therapeutic benefit. The present findings may contribute to the development of novel therapeutic strategies for pancreatic cancer.

The present study is an in vitro investigation focusing on the inhibition of pancreatic cancer cells compared to normal-fibroblast proliferation through the synergistic action of the three agents only on the pancreatic-cancer cells in contrast to normal cells. Further studies are needed to elucidate the underlying mechanisms, particularly regarding cell-cycle dynamics and autophagy. Although the present results suggest that the triple combination has minimal effects on normal cells, future clinical applications will require comprehensive in vivo and clinical evaluation to confirm the selective efficacy and safety of this approach.

Conclusion

The combination of rMETase, ivermectin, and chloroquine demonstrated selective synergistic efficacy against pancreatic cancer cells, which was not observed on normal cells. Furthermore, this combination therapy may be most effective when initiated early in the course of treatment. The present findings suggest the potential of this triple combination as a targeted therapeutic strategy for pancreatic cancer. However, further studies using in vivo models are necessary to evaluate the efficacy and safety of this combination treatment in selectively targeting pancreatic cancer.

Methionine restriction is effective because it targets a fundamental hallmark of cancer (3-9, 62-75).

Acknowledgements

This article is dedicated to the memory of A.R. Moossa, MD, Sun Lee, MD,, Professor Philip Miles, Richard W. Erbe, Professor Milton Plesur, MD, Professor Gordon H. Sato, Professor Li Jiaxi, Masaki Kitajima, MD, Joseph R. Bertino, MD, Shigeo Yagi, PhD, J.A.R Mead, PhD, Eugene P. Frenkel, 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, SL, KM, BMK, JSK, YM, 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 to declare in relation to this study.

  • Artificial Intelligence (AI) Disclosure

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

  • Received July 29, 2025.
  • Revision received August 18, 2025.
  • Accepted August 19, 2025.
  • Copyright © 2025 The Author(s). Published by the International Institute of Anticancer Research.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

References

  1. ↵
    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
  2. ↵
    1. Yang J,
    2. Xu R,
    3. Wang C,
    4. Qiu J,
    5. Ren B,
    6. You L
    : Early screening and diagnosis strategies of pancreatic cancer: a comprehensive review. Cancer Commun (Lond) 41(12): 1257-1274, 2021. DOI: 10.1002/cac2.12204
    OpenUrlCrossRefPubMed
  3. ↵
    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
  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 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 USA 79(14): 4248-4251, 1982. DOI: 10.1073/pnas.79.14.4248
    OpenUrlAbstract/FREE Full Text
  5. ↵
    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
  6. ↵
    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
    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
  7. ↵
    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): 1041-1046, 2010.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    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. 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. 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. Sato M,
    2. Sato T,
    3. Hozumi C,
    4. Han Q,
    5. Mizuta K,
    6. Morinaga S,
    7. Kang BM,
    8. Kobayashi N,
    9. Ichikawa Y,
    10. Nakajima A,
    11. Hoffman RM
    : [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. Anticancer Res 44(9): 3891-3898, 2024. DOI: 10.21873/anticanres.17216
    OpenUrlAbstract/FREE Full Text
  9. ↵
    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
    : Prostate cancer patient with lymph-node metastasis treated only with methionine restriction has stable disease for two years demonstrated with PET/CT and PSMA-PET scanning and PSA testing. Cancer Diagn Progn 5(1): 27-31, 2025. DOI: 10.21873/cdp.10408
    OpenUrlCrossRefPubMed
  10. ↵
    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
  11. ↵
    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
  12. ↵
    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
  13. ↵
    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
  14. ↵
    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
  15. ↵
    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
    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: 3397-3403, 2018. DOI: 10.3892/mmr.2017.8231
    OpenUrlCrossRef
  16. ↵
    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
  17. ↵
    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
  18. ↵
    1. Amaravadi RK,
    2. Kimmelman AC,
    3. Debnath J
    : Targeting autophagy in cancer: recent advances and future directions. Cancer Discov 9(9): 1167-1181, 2019. DOI: 10.1158/2159-8290.CD-19-0292
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Maes H,
    2. Kuchnio A,
    3. Peric A,
    4. Moens S,
    5. Nys K,
    6. De Bock K,
    7. Quaegebeur A,
    8. Schoors S,
    9. Georgiadou M,
    10. Wouters J,
    11. Vinckier S,
    12. Vankelecom H,
    13. Garmyn M,
    14. Vion AC,
    15. Radtke F,
    16. Boulanger C,
    17. Gerhardt H,
    18. Dejana E,
    19. Dewerchin M,
    20. Ghesquière B,
    21. Annaert W,
    22. Agostinis P,
    23. Carmeliet P
    : Tumor vessel normalization by chloroquine independent of autophagy. Cancer Cell 26(2): 190-206, 2014. DOI: 10.1016/j.ccr.2014.06.025
    OpenUrlCrossRefPubMed
  20. ↵
    1. Masud Alam M,
    2. Kariya R,
    3. Kawaguchi A,
    4. Matsuda K,
    5. Kudo E,
    6. Okada S
    : Inhibition of autophagy by chloroquine induces apoptosis in primary effusion lymphoma in vitro and in vivo through induction of endoplasmic reticulum stress. Apoptosis 21(10): 1191-1201, 2016. DOI: 10.1007/s10495-016-1277-7
    OpenUrlCrossRefPubMed
  21. ↵
    1. Harhaji-Trajkovic L,
    2. Arsikin K,
    3. Kravic-Stevovic T,
    4. Petricevic S,
    5. Tovilovic G,
    6. Pantovic A,
    7. Zogovic N,
    8. Ristic B,
    9. Janjetovic K,
    10. Bumbasirevic V,
    11. Trajkovic V
    : Chloroquine-mediated lysosomal dysfunction enhances the anticancer effect of nutrient deprivation. Pharm Res 29(8): 2249-2263, 2012. DOI: 10.1007/s11095-012-0753-1
    OpenUrlCrossRefPubMed
  22. ↵
    1. Seitz C,
    2. Hugle M,
    3. Cristofanon S,
    4. Tchoghandjian A,
    5. Fulda S
    : The dual PI3K/mTOR inhibitor NVP-BEZ235 and chloroquine synergize to trigger apoptosis via mitochondrial-lysosomal cross-talk. Int J Cancer 132(11): 2682-2693, 2013. DOI: 10.1002/ijc.27935
    OpenUrlCrossRefPubMed
  23. ↵
    1. Chen RJ,
    2. Lyu YJ,
    3. Chen YY,
    4. Lee YC,
    5. Pan MH,
    6. Ho YS,
    7. Wang YJ
    : Chloroquine potentiates the anticancer effect of pterostilbene on pancreatic cancer by inhibiting autophagy and downregulating the RAGE/STAT3 pathway. Molecules 26(21): 6741, 2021. DOI: 10.3390/molecules26216741
    OpenUrlCrossRefPubMed
    1. Samaras P,
    2. Tusup M,
    3. Nguyen-Kim TDL,
    4. Seifert B,
    5. Bachmann H,
    6. von Moos R,
    7. Knuth A,
    8. Pascolo S
    : Phase I study of a chloroquine–gemcitabine combination in patients with metastatic or unresectable pancreatic cancer. Cancer Chemother Pharmacol 80(5): 1005-1012, 2017. DOI: 10.1007/s00280-017-3446-y
    OpenUrlCrossRefPubMed
    1. Frieboes HB,
    2. Huang JS,
    3. Yin WC,
    4. McNally LR
    : Chloroquine-mediated cell death in metastatic pancreatic adenocarcinoma through inhibition of autophagy. JOP 15(2): 189-197, 2014. DOI: 10.6092/1590-8577/1900
    OpenUrlCrossRefPubMed
    1. Wang G,
    2. Chen S,
    3. Edwards H,
    4. Cui X,
    5. Cui L,
    6. Ge Y
    : Combination of chloroquine and GX15-070 (obatoclax) results in synergistic cytotoxicity against pancreatic cancer cells. Oncol Rep 32(6): 2789-2794, 2014. DOI: 10.3892/or.2014.3525
    OpenUrlCrossRefPubMed
  24. ↵
    1. Balic A,
    2. Sørensen MD,
    3. Trabulo SM,
    4. Sainz B Jr.,
    5. Cioffi M,
    6. Vieira CR,
    7. Miranda-Lorenzo I,
    8. Hidalgo M,
    9. Kleeff J,
    10. Erkan M,
    11. Heeschen C
    : Chloroquine targets pancreatic cancer stem cells via inhibition of CXCR4 and Hedgehog signaling. Mol Cancer Ther 13(7): 1758-1771, 2014. DOI: 10.1158/1535-7163.MCT-13-0948
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Asano Y,
    2. Han Q,
    3. Li S,
    4. Mizuta K,
    5. Kang BM,
    6. Kim JS,
    7. Miyashi Y,
    8. Yamamoto N,
    9. Hayashi K,
    10. Kimura H,
    11. Miwa S,
    12. Igarashi K,
    13. Higuchi T,
    14. Morinaga S,
    15. Tsuchiya H,
    16. Demura S,
    17. Hoffman RM
    : The combination of the autophagy inhibitor chloroquine and recombinant methioninase has selective synergistic efficacy on human colon cancer cells but not on normal human fibroblasts. Anticancer Res 45(7): 2825-2831, 2025. DOI: 10.21873/anticanres.17651
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Kim J,
    2. Han Q,
    3. Kang BM,
    4. Mizuta K,
    5. Asano Y,
    6. Bouvet M,
    7. Hoffman RM
    : The triple combination of recombinant methioninase, rapamycin, and chloroquine synergistically eradicates HCT116 colon cancer cells. Anticancer Res 45(7): 2773-2779, 2025. DOI: 10.21873/anticanres.17646
    OpenUrlAbstract/FREE Full Text
  27. ↵
    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
  28. ↵
    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
  29. ↵
    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
  30. ↵
    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. 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
    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. 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
  31. ↵
    1. Higuchi T,
    2. Oshiro H,
    3. Miyake K,
    4. Sugisawa N,
    5. Han Q,
    6. Tan Y,
    7. Park J,
    8. Zhang Z,
    9. Razmjooei S,
    10. Yamamoto N,
    11. Hayashi K,
    12. Kimura H,
    13. Miwa S,
    14. Igarashi K,
    15. Bouvet M,
    16. Chawla SP,
    17. Singh SR,
    18. Tsuchiya H,
    19. Hoffman RM
    : Oral recombinant methioninase, combined with oral caffeine and injected cisplatinum, overcome cisplatinum-resistance and regresses patient-derived orthotopic xenograft model of osteosarcoma. Anticancer Res 39(9): 4653-4657, 2019. DOI: 10.21873/anticanres.13646
    OpenUrlAbstract/FREE Full Text
  32. ↵
    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
  33. ↵
    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
  34. ↵
    1. Jiang L,
    2. Wang P,
    3. Sun YJ,
    4. Wu YJ
    : Ivermectin reverses the drug resistance in cancer cells through EGFR/ERK/Akt/NF-κB pathway. J Exp Clin Cancer Res 38(1): 265, 2019. DOI: 10.1186/s13046-019-1251-7
    OpenUrlCrossRefPubMed
  35. ↵
    1. Asano Y,
    2. Han Q,
    3. Li S,
    4. Mizuta K,
    5. Kang BM,
    6. Kim JS,
    7. Yamamoto N,
    8. Hayashi K,
    9. Kimura H,
    10. Miwa S,
    11. Igarashi K,
    12. Higuchi T,
    13. Morinaga S,
    14. Tsuchiya H,
    15. Demura S,
    16. Hoffman RM
    : Selective synergy of ivermectin combined with recombinant methioninase against colon-cancer cells in contrast to normal fibroblasts. Anticancer Res 45(6): 2257-2263, 2025. DOI: 10.21873/anticanres.17600
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Sasaki K,
    2. Tsuno NH,
    3. Sunami E,
    4. Tsurita G,
    5. Kawai K,
    6. Okaji Y,
    7. Nishikawa T,
    8. Shuno Y,
    9. Hongo K,
    10. Hiyoshi M,
    11. Kaneko M,
    12. Kitayama J,
    13. Takahashi K,
    14. Nagawa H
    : Chloroquine potentiates the anti-cancer effect of 5-fluorouracil on colon cancer cells. BMC Cancer 10: 370, 2010. DOI: 10.1186/1471-2407-10-370
    OpenUrlCrossRefPubMed
  37. ↵
    1. Choi JH,
    2. Yoon JS,
    3. Won YW,
    4. Park BB,
    5. Lee YY
    : Chloroquine enhances the chemotherapeutic activity of 5-fluorouracil in a colon cancer cell line via cell cycle alteration. APMIS 120(7): 597-604, 2012. DOI: 10.1111/j.1600-0463.2012.02876.x
    OpenUrlCrossRefPubMed
  38. ↵
    1. Shi S,
    2. Tan P,
    3. Yan B,
    4. Gao R,
    5. Zhao J,
    6. Wang J,
    7. Guo J,
    8. Li N,
    9. Ma Z
    : ER stress and autophagy are involved in the apoptosis induced by cisplatin in human lung cancer cells. Oncol Rep 35(5): 2606-2614, 2016. DOI: 10.3892/or.2016.4680
    OpenUrlCrossRefPubMed
  39. ↵
    1. Liu Y,
    2. Zhu Y,
    3. Gu L,
    4. Li K,
    5. Ma A,
    6. Liu L,
    7. Meng Y,
    8. Zhang J,
    9. Shen S,
    10. Shi Q,
    11. Liu D,
    12. Zhang X,
    13. Zhang S,
    14. Chai X,
    15. Gao P,
    16. Xing J,
    17. Wang Y,
    18. Chen H,
    19. Liu R,
    20. Du Q,
    21. Liu H,
    22. Dai L,
    23. Wang J
    : Chloroquine suppresses colorectal cancer progression via targeting CHKA and PFKM to inhibit the PI3K/AKT pathway and the Warburg effect. Int J Biol Sci 21(4): 1619-1631, 2025. DOI: 10.7150/ijbs.101921
    OpenUrlCrossRefPubMed
  40. ↵
    1. Kim J,
    2. Han Q,
    3. Kang BM,
    4. Mizuta K,
    5. Asano Y,
    6. Bouvet M,
    7. Hoffman RM
    : Combination of recombinant methioninase with rapamycin or chloroquine is synergistic to highly inhibit triple-negative breast cancer cells in vitro. Anticancer Res 45(5): 1853-1859, 2025. DOI: 10.21873/anticanres.17564
    OpenUrlAbstract/FREE Full Text
  41. ↵
    1. Morinaga S,
    2. Han Q,
    3. Mizuta K,
    4. Kang BM,
    5. Bouvet M,
    6. Yamamoto N,
    7. Hayashi K,
    8. Kimura H,
    9. Miwa S,
    10. Igarashi K,
    11. Higuchi T,
    12. Tsuchiya H,
    13. Demura S,
    14. Hoffman RM
    : Highly synergistic eradication of 143B osteosarcoma cells in vitro by the combination of recombinant methioninase, chloroquine, and rapamycin targeting methionine addiction, autophagy, and mTOR, respectively. Anticancer Res 45(3): 935-941, 2025. DOI: 10.21873/anticanres.17481
    OpenUrlAbstract/FREE Full Text
  42. ↵
    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
  43. ↵
    1. Xin L,
    2. Zhou LQ,
    3. Liu L,
    4. Yuan YW,
    5. Zhang HT,
    6. Zeng F
    : METase promotes cell autophagy via promoting SNHG5 and suppressing miR-20a in gastric cancer. Int J Biol Macromol 122: 1046-1052, 2019. DOI: 10.1016/j.ijbiomac.2018.09.051
    OpenUrlCrossRefPubMed
  44. ↵
    1. Xin L,
    2. Li SH,
    3. Liu C,
    4. Zeng F,
    5. Cao JQ,
    6. Zhou LQ,
    7. Zhou Q,
    8. Yuan YW
    : Methionine represses the autophagy of gastric cancer stem cells via promoting the methylation and phosphorylation of RAB37. Cell Cycle 19(20): 2644-2652, 2020. DOI: 10.1080/15384101.2020.1814044
    OpenUrlCrossRefPubMed
  45. ↵
    1. Ardjmand D,
    2. Sato M,
    3. Han Q,
    4. Kubota Y,
    5. Mizuta K,
    6. Morinaga S,
    7. Hoffman RM
    : Synergy of rapamycin and methioninase on colorectal cancer cells requires simultaneous and not sequential administration: implications for mTOR inhibition. Cancer Diagn Progn 4(4): 396-401, 2024. DOI: 10.21873/cdp.10338
    OpenUrlCrossRefPubMed
  46. ↵
    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. 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. Montalbano S,
    2. Raboni S,
    3. Sidoli S,
    4. Mozzarelli A,
    5. Bettati S,
    6. Buschini A
    : Post-translational modifications of histone variants in the absence and presence of a methionine-depleting enzyme in normal and cancer cells. Cancers (Basel) 15(2): 527, 2023. DOI: 10.3390/cancers15020527
    OpenUrlCrossRefPubMed
    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. 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. 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
    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. 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. 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. Han Q,
    4. Sugisawa N,
    5. Sun Y,
    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. Andronicos A,
    2. Yoneda KC,
    3. Lin D-W,
    4. Law FV,
    5. Bae H,
    6. Basirattalab A,
    7. Graham NA,
    8. Jang C,
    9. Kaiser P
    : Carboxy-methylation of the catalytic subunit of protein phosphatase 2A (PP2Ac) integrates methionine availability with methionine addicted cancer cell proliferation. Biomolecules 15(9): 1210, 2025. DOI: 10.3390/biom15091210
    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
  47. ↵
    1. Hoffman RM,
    2. Jacobsen SJ,
    3. Erbe RW
    : Reversion to methionine independence in simian virus 40-transformed human and malignant rat fibroblasts is associated with altered ploidy and altered properties of transformation. Proc Natl Acad Sci USA 76(3): 1313-1317, 1979. DOI: 10.1073/pnas.76.3.1313
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

Anticancer Research: 45 (11)
Anticancer Research
Vol. 45, Issue 11
November 2025
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Ed Board (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.
Triple Combination of Recombinant Methioninase and the Anti-parasitic Drugs Ivermectin, and Chloroquine Selectively Eradicates Pancreatic Cancer Cells While Sparing 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.
2 + 5 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Triple Combination of Recombinant Methioninase and the Anti-parasitic Drugs Ivermectin, and Chloroquine Selectively Eradicates Pancreatic Cancer Cells While Sparing Normal Fibroblasts
YOHEI ASANO, QINGHONG HAN, SHUKUAN LI, KOHEI MIZUTA, BYUNG MO KANG, JIN SOO KIM, YUTA MIYASHI, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, SEI MORINAGA, HIROYUKI TSUCHIYA, SATORU DEMURA, ROBERT M. HOFFMAN
Anticancer Research Nov 2025, 45 (11) 4791-4802; DOI: 10.21873/anticanres.17828

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Triple Combination of Recombinant Methioninase and the Anti-parasitic Drugs Ivermectin, and Chloroquine Selectively Eradicates Pancreatic Cancer Cells While Sparing Normal Fibroblasts
YOHEI ASANO, QINGHONG HAN, SHUKUAN LI, KOHEI MIZUTA, BYUNG MO KANG, JIN SOO KIM, YUTA MIYASHI, NORIO YAMAMOTO, KATSUHIRO HAYASHI, HIROAKI KIMURA, SHINJI MIWA, KENTARO IGARASHI, TAKASHI HIGUCHI, SEI MORINAGA, HIROYUKI TSUCHIYA, SATORU DEMURA, ROBERT M. HOFFMAN
Anticancer Research Nov 2025, 45 (11) 4791-4802; DOI: 10.21873/anticanres.17828
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

  • Simultaneous Targeting of Multiple Hallmarks of Cancer With Recombinant Methioninase, Rapamycin and Chloroquine Is Specific and Synergistic to MiaPaCa-2 Pancreatic-Cancer Cells in Contrast to Hs-27 Normal Fibroblasts
  • Contribution of Xeroderma Pigmentosum Complementation Group C Genotypes to Colorectal Cancer in Taiwanese
  • Phenotypic Alteration and Suppression of Cytotoxicity of Decidual NK Cells After Co-culturing With Different Trophoblastic Cell Lines
Show more Experimental Studies

Similar Articles

Keywords

  • Recombinant methioninase (rMETase)
  • ivermectin
  • chloroquine
  • combination treatment
  • synergy
  • pancreatic cancer
  • normal fibroblasts
  • methionine addiction
  • Hoffman effect
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire