Open Access

Synergy of Combining Methionine Restriction and Chemotherapy: The Disruptive Next Generation of Cancer Treatment

KUBOTA YUTARO 1 2 3
HAN QINGHONG 1
AOKI YUSUKE 1 2
MASAKI NORIYUKI 1 2
OBARA KOYA 1 2
HAMADA KAZUYUKI 1 2 3
HOZUMI CHIHIRO 4
WONG ANDREW C.W. 5
BOUVET MICHAEL 2
TSUNODA TAKUYA 3
  &  
HOFFMAN ROBERT M. 1 2

1AntiCancer Inc., San Diego, CA, U.S.A.

2Department of Surgery, University of California, San Diego, CA, U.S.A.

3Division of Internal Medicine, Department of Medical Oncology, Showa University School of Medicine, Tokyo, Japan

4AntiCancer Japan Inc., Narita, Japan

5Clinic of Advanced Cancer Treatment & Regeneration Medicine, Tokyo, Japan

Cancer Diagnosis & Prognosis May-June; 3(3): 272-281 DOI: 10.21873/cdp.10212
Received 16 January 2023 | Revised 03 December 2024 | Accepted 06 March 2023
Corresponding author
Robert M. Hoffman, Ph.D., AntiCancer Inc, 7917 Ostrow St, Suite B, San Diego, CA, 92111, U.S.A. Tel: +1 6198852284, email: all@anticancer.com

Abstract

All cancer cell types are methionine-addicted, which is termed the Hoffman effect. Cancer cells, unlike normal cells, cannot survive without large amount of methionine. In general, when methionine is depleted, both normal cells and cancer cells synthesize methionine from homocysteine, but cancer cells consume large amounts of methionine and they cannot survive without exogenous methionine. For this reason, methionine restriction has been shown to be effective against many cancers in vitro and in vivo. Methionine restriction arrests cancer cells in the S/G2-phase of the cell cycle. Cytotoxic agents that act in the S/G2-phase are highly effective when used in combination with methionine restriction due to the cancer cells being trapped in S/G2-phase, unlike normal cells which arrest in G1/G0-phase. Combining methionine restriction and chemotherapeutic drugs for cancer treatment is termed the Hoffman protocol. The efficacy of many cytotoxic agents and molecular-targeted drugs in combination with methionine restriction has been demonstrated. The most effective method of methionine restriction is the administration of recombinant methioninase (rMETase), which degrades methionine. The efficacy of rMETase has been reported in mice and human patients by oral administration. The present review describes studies on anticancer drugs that showed synergistic efficacy in combination with methionine restriction, including rMETase administration. It is proposed that the next disruptive generation of cancer chemotherapy should employ current therapy in combination with methionine restriction for all cancer types.
Keywords: Methionine restriction, Hoffman effect, cancer, chemotherapy, recombinant methioninase (rMETase), combination therapy, synergy, Hoffman protocol, review

Methionine dependence of cancer was discovered by Sugimura et al. in 1959 (1) when it was observed that methionine-depleted rat chow slowed cancer growth in rats more than chow depleted of other amino acids. Fourteen years later, Chello and Bertino (2) found that leukemia and lymphoma cells could not grow in culture, while normal cells could grow, when methionine was replaced by its immediate precursor, homocysteine.

In 1976, we found that cancer cells make normal or large amount of methionine, but still usually require an exogenous source of methionine (3). The conclusion was that cancer cells are addicted to methionine. Wang et al. 43 years later, confirmed that cancer cells are addicted to methionine, especially tumor-initiating or stem cells (4).

We originally showed that cancer cells were methionine-addicted, at least in part due to elevated transmethylation reactions (5), with histone lysine marks being over-methylated in cancer (6–9). Rare revertant of cancer cells that have regained methionine independence, have reduced levels of transmethylation (10,11), have lost their malignancy (7-9), and do not necessarily increase their ability to make methionine from homocysteine (12).

The methionine addiction of cancer is known as the Hoffman effect. Comparison of [11C]methionine positron emission tomography (PET) and [18F]deoxyglucose PET has shown that the Hoffman effect is more pronounced than the Warburg effect of cancer addiction to glucose (13).

We first observed selective cell-cycle arrest of cancer cells in S/G2-phase when they are depleted of methionine (14). The S/G2-phase of the cell cycle is the vulnerable point of cytotoxic chemotherapy, resulting in the possibility of synergy. We first observed this synergy almost 40 years ago when normal and cancer cells were co-cultured with the combination of methionine restriction and chemotherapy, which could selectively eliminate all the cancer cells, leaving healthy intact normal cells (15). Subsequently, we have shown synergy of all major types of chemotherapy and methionine restriction, including with recombinant methioninase (rMETase). We believe that the next disruptive generation of cancer treatment will combine methionine restriction and current chemotherapy.

Doxorubicin (DOX) Synergy With Methionine Restriction

The first chemotherapeutic drug that showed a synergistic effect with methionine restriction was doxorubicin (DOX). As mentioned above, cancer cells become reversibly blocked in the late S/G2-phase under methionine depletion (14). DOX blocks topoisomerase 2 and targets S-phase and was predicted to be synergistic with methionine restriction. Stern et al. (15) demonstrated the synergistic effect of methionine restriction and DOX using sarcoma, prostate cancer, lung cancer, and breast cancer cell lines co-cultured with normal fibroblasts. Homocysteine was substituted for methionine in the culture medium and DOX was then added. After administering DOX to the cancer cells that had been arrested in S/G2-phase by methionine restriction, methionine was added to stimulate the cancer cells to synchronously resume cycling and then vincristine was added, which acts in M-phase to kill the cancer cells entering mitosis. Normal cells were protected because they arrest in G0-phase when methionine-depleted. This disruptive strategy resulted in selectively eliminating all the cancer cells from the co-culture, leaving a healthy culture of normal fibroblasts (15) (Table I).

Gupta et al. showed the efficacy of the combination of DOX and methioninase, using a recombinant adenovirus (Ad-MET) which produced methioninase, on the human lung-cancer cell line H460 (16). Selenomethionine (SeMET), which is degraded by methioninase and results in the highly toxic methylselonol that has a strong bystander effect, was also added as a pro-drug. The combination treatment of DOX, Ad-MET, and SeMET inhibited tumor growth more than the combination of Ad-MET and SeMET.

Recently, the synergistic efficacy of the combination of DOX and recombinant methioninase (rMETase) (oral or intraperitoneal dosing) was reported in synovial sarcoma (17,18) and undifferentiated spindle-cell carcinoma (19,20) in patient-derived orthotopic xenograft (PDOX) mouse models (Table I).

A patient with invasive lobular carcinoma of the breast who received DOX and cyclophosphamide combined with rMETase and a low-methionine diet had her axillary-lymph-node metastasis eliminated (21) (Table I).

5-Fluorouracil (5-FU) Synergy With Methionine Restriction

5-Fluorouracil (5-FU) is an anti-metabolite that targets cells in the S-phase of the cell cycle similar to DOX. Hoshiya et al. (22) originally showed the synergistic efficacy of the combination of methionine restriction and 5-FU using the human gastric-cancer cell-line (SC-1-NU) xenograft mouse model. This study demonstrated that methionine restriction enhanced the antitumor activity of 5-FU by approximately two-fold. Hoshiya et al. also showed that methionine restriction increased intra-tumoral thymidylate-synthase (TS) inhibition by 5-FU. Recently, Gao et al. (23) showed that methionine restriction combined with 5-FU enhanced the treatment response in RAS-driven colorectal cancer in patient-derived xenograft (PDX) models, confirming the original result of Hoshiya et al. (22) (Table I).

A clinical trial for gastric cancer patients was performed using 5-FU combined with methionine-free total parenteral nutrition (TPN) (24). Fourteen patients with gastric cancer who had stenosis or obstruction of the gastric canal were registered in this trial. The patients were randomly allocated into two groups, 5-FU and methionine-free TPN, or 5-FU and normal (methionine-containing) TPN. All patients received surgery after 7 days of 5-FU and TPN. The specimens in the methionine-free TPN group showed extensive degeneration of cancer. Also, the TS activity was decreased in the tumor in the methionine-free TPN group (Table I).

Subsequently, the synergistic efficacy of the combination of 5-FU and rMETase was shown using a xenograft model of Lewis lung carcinoma cells, including extended survival (25). Machover et al. demonstrated the efficacy of adding folinic acid to 5-FU and rMETase on the CCRF-CEM human T-lymphoblastic leukemia cell line in vitro (26). The synergistic efficacy of 5-FU and oral rMETase was observed in colorectal cancer, poorly-differentiated gastric cancer, and colon-cancer peritoneal-carcinomatosis mouse models (27–29) (Table I).

Lu et al. (30) showed that methionine restriction combined with 5-FU reduced 5,10-methylene-tetrahydrofolate levels by 75% and selectively inhibited TS activity in PC-3 human prostate-cancer cells. Reduction of 5,10-methylene-tetrahydrofolate decreased the level of 5-methyl-tetra-hydrofolate, which is necessary for methionine synthesis. 5,10-methylene-tetrahydrofolate depletion and decreased TS activity, due to methionine restriction, resulted in synergistic efficacy with 5-FU (30) (Table I).

Gemcitabine (GEM) Synergy With Methionine Restriction

Gemcitabine (GEM) is also classified as an anti-metabolite targeting cells in S-phase. The synergistic efficacy of GEM and rMETase (intraperitoneal and oral dosing) was reported in PDOX and orthotopic cell-line mouse models of pancreatic cancer (31,32). These studies showed that the combination of GEM and rMETase was synergistically effective for GEM-resistant pancreatic cancer. GEM resistance can be overcome by using methionine restriction in combination with the drug to which the cancer cells are resistant (Table I).

Methotrexate (MTX) Synergy With Methionine Restriction

MTX is also classified as an anti-metabolite. MTX inhibits dihydrofolate reductase (DHFR) and methylene-tetra-hydrofolate reductase (MTHFR), which decrease the level of 5-methyl-tetrahydrofolate that adds a methyl group to homocysteine, and therefore blocks endogenous methionine production (33). MTX also inhibits methionine S-adenosyltransferase (MAT). MTX, combined with rMETase, was therefore synergistic on an MTX-resistant osteosarcoma PDOX model (33) (Table I).

Platinum-based Chemotherapy Synergy With Methionine Restriction

Platinum agents, such as cisplatinum and oxaliplatinum bind to nuclear DNA and subsequently interfere with DNA replication. Hoshiya et al. originally demonstrated the synergistic efficacy of cisplatinum and methionine restriction in vitro and on a xenograft mouse model utilizing the human breast cancer cell line MX-1 (34). In addition, cisplatinum combined with rMETase showed efficacy for colon cancer cell lines (Colo205, SW620, HCT15, and HT29) xenograft mouse models (35), and osteosarcoma PDOX and orthotopic xenograft mouse models (36–38), and a bladder-cancer orthotopic mouse model (39).

Oxaliplatinum is used for colon cancer combined with 5-FU because its monotherapy is not effective in clinical settings. This combination is termed FOLFOX. FOLFOX showed synergistic efficacy combined with oral rMETase in a colon-cancer PDOX mouse model (27) and orthotopic xenograft mouse model (29).

A clinical trial was performed using the combination of a low-methionine diet and FOLFOX. This trial included 11 patients with unresectable colorectal cancer. From the start of chemotherapy, all patients were on a methionine-free diet for three days. Of the 4 patients evaluable for response, 3 experienced a partial response, and 1 patient had stable disease (40). Recently, a case of a patient with stage IV pancreatic cancer who received oxaliplatinum with 5-FU and irinotecan, which is termed FOLFIRINOX, combined with a low-methionine diet and rMETase, has 18 months of stable disease, a result found in only 5% of stage IV pancreatic cancer patient (41) (Table I).

Alkylating Agents Synergy With Methionine Restriction

Kokkinakis et al. showed the synergistic efficacy of alkylating agents [carmustine (BCNU) and temozolomide] and methionine restriction in brain-cancer xenograft mouse models, using rMETase and low-methionine mouse chow, including Daoy (medulloblastoma), SWB77 (glioblastoma), and D-54 (glioblastoma) (42). In general, glioblastomas with MGMT activation are resistant to temozolomide. However, methionine restriction decreased MGMT activation (42). Therefore, temozolomide with methionine restriction is effective against glioblastoma, even if MGMT is activated. The synergistic efficacy of temozolomide and intraperitoneal rMETase was also reported in a BRAF mutant melanoma PDOX mouse model (43).

The combination of cystemustine and a methionine- restricted diet was examined in human phase 1 and 2 trials for melanoma and high-grade glioma patients (44,45). In the phase 2 trial, the period of methionine restriction was set to only one day for each two-week cystemustine cycle, based on the results of the phase 1 trial. The results of these trials showed that this combination of cystemustine and methionine restriction has less toxicity, but no efficacy, compared to historical controls. One-day methionine restriction is too short a period for depleting methionine (Table I).

Tubulin-targeting Drugs Synergy With Methionine Restriction

Taxanes, such as paclitaxel and docetaxel, as well as eribulin, a halichondrine, affect the M-phase of the cell cycle by interfering with microtubules. Cancer cells that escape the S/G2 cell-cycle block induced by methionine restriction are then killed by tubulin-targeting drugs as they enter M-phase. Paclitaxel for clear-cell ovarian cancer (46), eribulin for triple-negative cancer (47), and docetaxel for osteosarcoma (48) were synergistic with oral rMETase in PDOX mouse models (Table I).

Tamoxifen Synergy With Methionine Restriction

Tamoxifen, a hormone analog that inhibits the estrogen receptor, showed synergistic efficacy with rMETase in a breast-cancer orthotopic xenograft mouse model (MCF-7) (49). The combination of rMETase and tamoxifen increased caspase-3 and -8 expression, indicating apoptosis. The mechanism of the synergy of this combination is not clear (Table I).

Palbociclib Synergy With Methionine Restriction

Palbociclib is a cyclin-dependent kinase (CDK)-4 and -6 inhibitor (50). This blockade inhibits the progress of the cell cycle from the G1-phase to the S-phase. In a PDOX mouse model of DOX-resistant dedifferentiated liposarcoma, the synergistic efficacy of palbociclib and rMETase was demonstrated (50). These results show that the double blockade of the cell cycle (phases G1 to S and S to G2) is effective against cancer cells (Table I).

DNA-methylation-inhibitor Synergy With Methionine Restriction

Azacytidine and decitabine are classified as hypo-methylating agents. They prevent the methylation of the cytosines in DNA by inhibiting DNA methyltransferase (51). Methionine restriction decreases S-adenosylmethionine (SAM) (52), which is the only methyl-group provider for DNA, RNA, and histone methylation. In osteosarcoma and soft-tissue sarcoma PDOX mouse models, the combination of azacytidine or decitabine and rMETase was synergistically effective (51,53). Another PDOX mouse model study using pancreatic cancer demonstrated the synergistic efficacy of rMETase, azacytidine, and cycloleucine, which is a specific inhibitor of SAM synthesis (54). Further study is needed to investigate the effect of methionine restriction on DNA methylation (Table I).

Rapamycin Synergy With Methionine Restriction

Rapamycin targets mTOR kinase and inhibits the PI3K/AKT signaling pathway (55). The synergistic efficacy of rapamycin and oral rMETase was reported in an osteosarcoma- of-the-breast PDOX mouse model (56) (Table I).

Targeting TRAIL Receptor-2 Synergy With Methionine Restriction

Tigatuzumab and lexatumumab target TNF-related apoptosis-induced ligand receptor-2 (TRAIL-R2) (57,58). Methionine restriction increased TRAIL-R2 expression in cancer cells. Lexatumumab inhibited triple-negative breast cancer in vitro and in mice with low-methionine medium or diet. Tigatuzumab combined with oral rMETase showed synergy on pancreatic- cancer orthotopic xenograft mouse models (MIA PaCa-2 and BxPC-3) (58) (Table I).

Discussion

It was first shown by Sugimura et al. in 1959 (1) that cancers are methionine-dependent and subsequently Hoffman and Erbe showed cancers are methionine-addicted in 1976 (3). It was then shown that all cancers are methionine addicted (59,60). As described in the present report, many mouse experiments and human studies have shown the synergy of different anticancer drugs with methionine restriction. Anti-metabolites, DOX, alkylating agents, and platinum drugs target cells in S/G2-phase where cancer cells are selectively blocked by methionine restriction (14,28,61). In addition, methotrexate and 5-FU target folate metabolism, and therefore decrease the ability of cells to synthesize methionine, and show synergistic efficacy with methionine restriction. Methotrexate was shown to increase histone-lysine methylation in cancer cells, which alters cell programming (62). Other cytotoxic and molecular-targeting agents such as taxanes, tamoxifen, palbociclib, azacytidine, rapamycin, and tigatuzumab are also synergistic with methionine restriction.

In an emerging series of human studies (21,41), rMETase appears synergistic with chemotherapy. No side effects related to methionine restriction have been shown in either mouse or human studies.

The next disruptive generation of cancer treatment will be based on combining methionine restriction with current chemotherapy. which is termed the Hoffman protocol (56).

It should be noted that methionine addicted cancer cells are also addicted to folate (63) which contributes to the efficacy of anti-folate chemotherapy (33).

Despite overwhelming evidence that cancer cells express high levels of methionine synthase (3,4,64,65), misinformation published 50 years ago that methionine dependence of cancer is due to depleted methionine synthase (66,67) still persists (68).

Very recently a paper published in Nature (69) showed research is becoming less disruptive. This seems not to be the case for the next generation of cancer chemotherapy based on methionine addiction of cancer (3-11,13,52,70-73).

Emerging evidence suggests that methionine restriction sensitizes cancer cells to pro-oxidants (Table I) (74).

Conflicts of Interest

The Authors declare no competing interests regarding this work.

Authors’ Contributions

YK and RMH wrote the article. QH provided the recombinant methioninase. YA, NM, KO, KH, CH, AW, MB, and TT reviewed the article.

Acknowledgements

This paper is dedicated to the memory of A. R. Moossa, MD, Sun Lee, MD, Professor Li Jiaxi, Masaki Kitajima, MD, Shigeo Yagi, PhD, Jack Geller, MD, Joseph R. Bertino, MD, and J.A.R. Mead PhD. The Robert M. Hoffman Foundation for cancer research provided funds for this study.

References

1 Sugimura T Birnbaum SM Winitz M & Greenstein JP Quantitative nutritional studies with water-soluble, chemically defined diets. VIII. The forced feeding of diets each lacking in one essential amino acid. Arch Biochem Biophys. 81(2) 448 - 455 1959. PMID: 13638009. DOI: 10.1016/0003-9861(59)90225-5
2 Chello PL & Bertino JR Dependence of 5-methyltetrahydrofolate utilization by L5178Y murine leukemia cells in vitro on the presence of hydroxycobalamin and transcobalamin II. Cancer Res. 33(8) 1898 - 1904 1973. PMID: 4737200.
Pubmed |
3 Hoffman RM & 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. PMID: 179090. DOI: 10.1073/pnas.73.5.1523
4 Wang Z Yip LY Lee JHJ Wu Z Chew HY Chong PKW Teo CC Ang HY Peh KLE Yuan J Ma S Choo LSK Basri N Jiang X Yu Q Hillmer AM Lim WT Lim TKH Takano A Tan EH Tan DSW Ho YS Lim B & Tam WL Methionine is a metabolic dependency of tumor-initiating cells. Nat Med. 25(5) 825 - 837 2019. PMID: 31061538. DOI: 10.1038/s41591-019-0423-5
5 Stern PH & Hoffman RM Elevated overall rates of transmethylation in cell lines from diverse human tumors. In Vitro. 20(8) 663 - 670 1984. PMID: 6500606. DOI: 10.1007/BF02619617
6 Yamamoto J Han Q Inubushi S Sugisawa N Hamada K Nishino H Miyake K Kumamoto T Matsuyama R Bouvet M Endo I & 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. PMID: 33019978. DOI: 10.1016/j.bbrc.2020.09.108
7 Yamamoto J Aoki Y Han Q Sugisawa N Sun YU Hamada K Nishino H Inubushi S Miyake K Matsuyama R Bouvet M Endo I & 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. PMID: 33517268. DOI: 10.21873/anticanres.14815
8 Yamamoto J Aoki Y Inubushi S Han Q Hamada K Tashiro Y Miyake K Matsuyama R Bouvet M Clarke SG Endo I & 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. PMID: 34949655. DOI: 10.21873/cgp.20299
9 Aoki Y Han Q Tome Y Yamamoto J Kubota Y Masaki N Obara K Hamada K Wang JD Inubushi S Bouvet M Clarke SG Nishida K & 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. PMID: 36408173. DOI: 10.3389/fonc.2022.1009548
10 Hoffman RM Jacobsen SJ & 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. PMID: 220612. DOI: 10.1073/pnas.76.3.1313
11 Judde JG Ellis M & Frost P Biochemical analysis of the role of transmethylation in the methionine dependence of tumor cells. Cancer Res. 49(17) 4859 - 4865 1989. PMID: 2503245.
Pubmed |
12 Hoffman RM Jacobsen SJ & Erbe RW Reversion to methionine independence by malignant rat and SV40-transformed human fibroblasts. Biochem Biophys Res Commun. 82(1) 228 - 234 1978. PMID: 208554. DOI: 10.1016/0006-291x(78)90600-9
13 Kubota Y Sato T Hozumi C Han Q Aoki Y Masaki N Obara K Tsunoda T & Hoffman RM Superiority of [11C]methionine over [18F]deoxyglucose for PET imaging of multiple cancer types due to the methionine addiction of cancer. Int J Mol Sci. 24(3) 1935 2023. PMID: 36768257. DOI: 10.3390/ijms24031935
14 Hoffman RM & Jacobsen SJ Reversible growth arrest in simian virus 40-transformed human fibroblasts. Proc Natl Acad Sci USA. 77(12) 7306 - 7310 1980. PMID: 6261250. DOI: 10.1073/pnas.77.12.7306
15 Stern PH & 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. PMID: 3457200. DOI: 10.1093/jnci/76.4.629
16 Gupta A Miki K Xu M Yamamoto N Moossa AR & Hoffman RM Combination efficacy of doxorubicin and adenoviral methioninase gene therapy with prodrug selenomethionine. Anticancer Res. 23(2B) 1181 - 1188 2003. PMID: 12820369.
Pubmed |
17 Igarashi K Kawaguchi K Li S Han Q Tan Y Gainor E Kiyuna T Miyake K Miyake M Higuchi T Oshiro H Singh AS Eckardt MA Nelson SD Russell TA Dry SM Li Y Yamamoto N Hayashi K Kimura H Miwa S Tsuchiya H Eilber FC & Hoffman RM Recombinant methioninase combined with doxorubicin (DOX) regresses a DOX-resistant synovial sarcoma in a patient-derived orthotopic xenograft (PDOX) mouse model. Oncotarget. 9(27) 19263 - 19272 2018. PMID: 29721200.
Pubmed |
18 Higuchi T Kawaguchi K Miyake K Han Q Tan Y Oshiro H Sugisawa N Zhang Z Razmjooei S Yamamoto N Hayashi K Kimura H Miwa S Igarashi K Chawla SP Singh AS Eilber FC Singh SR Tsuchiya H & Hoffman RM Oral recombinant methioninase combined with caffeine and doxorubicin induced regression of a doxorubicin-resistant synovial sarcoma in a PDOX mouse model. Anticancer Res. 38(10) 5639 - 5644 2018. PMID: 30275182. DOI: 10.21873/anticanres.12899
19 Igarashi K Kawaguchi K Li S Han Q Tan Y Murakami T Kiyuna T Miyake K Miyake M Singh AS Eckardt MA Nelson SD Russell TA Dry SM Li Y Yamamoto N Hayashi K Kimura H Miwa S Tsuchiya H Singh SR Eilber FC & Hoffman RM Recombinant methioninase in combination with doxorubicin (DOX) overcomes first-line DOX resistance in a patient-derived orthotopic xenograft nude-mouse model of undifferentiated spindle-cell sarcoma. Cancer Lett. 417 168 - 173 2018. PMID: 29306021. DOI: 10.1016/j.canlet.2017.12.028
20 Igarashi K Li S Han Q Tan Y Kawaguchi K Murakami T Kiyuna T Miyake K Li Y Nelson SD Dry SM Singh AS Elliott IA Russell TA Eckardt MA Yamamoto N Hayashi K Kimura H Miwa S Tsuchiya H Eilber FC & Hoffman RM Growth of doxorubicin-resistant undifferentiated spindle-cell sarcoma PDOX is arrested by metabolic targeting with recombinant methioninase. J Cell Biochem. 119(4) 3537 - 3544 2018. PMID: 29143983. DOI: 10.1002/jcb.26527
21 Kubota Y Han Q Masaki N Hozumi C Hamada K Aoki Y Obara K Tsunoda T & 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. PMID: 36456116. DOI: 10.21873/anticanres.16089
22 Hoshiya Y Kubota T Inada T Kitajima M & Hoffman RM Methionine-depletion modulates the efficacy of 5-fluorouracil in human gastric cancer in nude mice. Anticancer Res. 17(6D) 4371 - 4375 1997. PMID: 9494535.
Pubmed |
23 Gao X Sanderson SM Dai Z Reid MA Cooper DE Lu M Richie JP Jr Ciccarella A Calcagnotto A Mikhael PG Mentch SJ Liu J Ables G Kirsch DG Hsu DS Nichenametla SN & Locasale JW Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature. 572(7769) 397 - 401 2019. PMID: 31367041. DOI: 10.1038/s41586-019-1437-3
24 Goseki N Yamazaki S Shimojyu K Kando F Maruyama M Endo M Koike M & Takahashi H Synergistic effect of methionine-depleting total parenteral nutrition with 5-fluorouracil on human gastric cancer: a randomized, prospective clinical trial. Jpn J Cancer Res. 86(5) 484 - 489 1995. PMID: 7790321. DOI: 10.1111/j.1349-7006.1995.tb03082.x
25 Yoshioka T Wada T Uchida N Maki H Yoshida H Ide N Kasai H Hojo K Shono K Maekawa R Yagi S Hoffman RM & Sugita K Anticancer efficacy in vivo and in vitro, synergy with 5-fluorouracil, and safety of recombinant methioninase. Cancer Res. 58(12) 2583 - 2587 1998. PMID: 9635582.
Pubmed |
26 Machover D Zittoun J Broët P Metzger G Orrico M Goldschmidt E Schilf A Tonetti C Tan Y Delmas-Marsalet B Luccioni C Falissard B & Hoffman RM Cytotoxic synergism of methioninase in combination with 5-fluorouracil and folinic acid. Biochem Pharmacol. 61(7) 867 - 876 2001. PMID: 11274973. DOI: 10.1016/s0006-2952(01)00560-3
27 Oshiro H Tome Y Kiyuna T Yoon SN Lwin TM Han Q Tan Y Miyake K Higuchi T Sugisawa N Katsuya Y Park JH Zang Z Razmjooei S Bouvet M Clary B Singh SR Kanaya F Nishida K & 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. PMID: 31519565. DOI: 10.21873/anticanres.13648
28 Miyake M Miyake K Han Q Igarashi K Kawaguchi K Barangi M Kiyuna T Sugisawa N Higuchi T Oshiro H Zhang Z Razmjooei S Bouvet M Endo I & Hoffman RM Synergy of oral recombinant methioninase (rMETase) and 5-fluorouracil on poorly differentiated gastric cancer. Biochem Biophys Res Commun. 643 48 - 54 2023. PMID: 36586158. DOI: 10.1016/j.bbrc.2022.12.062
29 Kim MJ Han Q Bouvet M Hoffman RM & 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. PMID: 36585181. DOI: 10.21873/anticanres.16129
30 Lu S Chen GL Ren C Kwabi-Addo B & Epner DE Methionine restriction selectively targets thymidylate synthase in prostate cancer cells. Biochem Pharmacol. 66(5) 791 - 800 2003. PMID: 12948860. DOI: 10.1016/s0006-2952(03)00406-4
31 Kawaguchi K Miyake K Han Q Li S Tan Y Igarashi K Lwin TM Higuchi T Kiyuna T Miyake M Oshiro H Bouvet M Unno M & Hoffman RM Targeting altered cancer methionine metabolism with recombinant methioninase (rMETase) overcomes partial gemcitabine-resistance and regresses a patient-derived orthotopic xenograft (PDOX) nude mouse model of pancreatic cancer. Cell Cycle. 17(7) 868 - 873 2018. PMID: 29623758. DOI: 10.1080/15384101.2018.1445907
32 Kawaguchi K Miyake K Han Q Li S Tan Y Igarashi K Kiyuna T Miyake M Higuchi T Oshiro H Zhang Z Razmjooei S Wangsiricharoen S Bouvet M Singh SR Unno M & Hoffman RM Oral recombinant methioninase (o-rMETase) is superior to injectable rMETase and overcomes acquired gemcitabine resistance in pancreatic cancer. Cancer Lett. 432 251 - 259 2018. PMID: 29928962. DOI: 10.1016/j.canlet.2018.06.016
33 Aoki Y Tome Y Han Q Yamamoto J Hamada K Masaki N Kubota Y Bouvet M Nishida K & Hoffman RM Oral-recombinant methioninase converts an osteosarcoma from methotrexate-resistant to -sensitive in a patient-derived orthotopic-xenograft (PDOX) mouse model. Anticancer Res. 42(2) 731 - 737 2022. PMID: 35093871. DOI: 10.21873/anticanres.15531
34 Hoshiya Y Kubota T Matsuzaki SW Kitajima M & Hoffman RM Methionine starvation modulates the efficacy of cisplatin on human breast cancer in nude mice. Anticancer Res. 16(6B) 3515 - 3517 1996. PMID: 9042214.
Pubmed |
35 Tan Y Sun X Xu M Tan X Sasson A Rashidi B Han Q Tan X Wang X An Z Sun FX & Hoffman RM Efficacy of recombinant methioninase in combination with cisplatin on human colon tumors in nude mice. Clin Cancer Res. 5(8) 2157 - 2163 1999. PMID: 10473100.
Pubmed |
36 Igarashi K Kawaguchi K Kiyuna T Miyake K Miyake M Li S Han Q Tan Y Zhao M Li Y Nelson SD Dry SM Singh AS Elliott IA Russell TA Eckardt MA Yamamoto N Hayashi K Kimura H Miwa S Tsuchiya H Eilber FC & Hoffman RM Tumor-targeting Salmonella typhimurium A1-R combined with recombinant methioninase and cisplatinum eradicates an osteosarcoma cisplatinum-resistant lung metastasis in a patient-derived orthotopic xenograft (PDOX) mouse model: decoy, trap and kill chemotherapy moves toward the clinic. Cell Cycle. 17(6) 801 - 809 2018. PMID: 29374999. DOI: 10.1080/15384101.2018.1431596
37 Higuchi T Oshiro H Miyake K Sugisawa N Han Q Tan Y Park J Zhang Z Razmjooei S Yamamoto N Hayashi K Kimura H Miwa S Igarashi K Bouvet M Chawla SP Singh SR Tsuchiya H & 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. PMID: 31519563. DOI: 10.21873/anticanres.13646
38 Masaki N Han Q Wu NF Samonte C Wu J Hozumi C Obara K Kubota Y Aoki Y Miyazaki J & Hoffman RM Oral-recombinant methioninase lowers the effective dose and eliminates toxicity of cisplatinum for primary osteosarcoma of the mammary gland in a patient-derived orthotopic xenograft mouse model. In Vivo. 36(6) 2598 - 2603 2022. PMID: 36309364. DOI: 10.21873/invivo.12994
39 Sun YU Nishino H Sugisawa N Yamamoto J Hamada K Zhu G Lim HI & Hoffman RM Oral recombinant methioninase sensitizes a bladder cancer orthotopic xenograft mouse model to low-dose cisplatinum and prevents metastasis. Anticancer Res. 40(11) 6083 - 6091 2020. PMID: 33109546. DOI: 10.21873/anticanres.14629
40 Durando X Farges MC Buc E Abrial C Petorin-Lesens C Gillet B Vasson MP Pezet D Chollet P & Thivat E Dietary methionine restriction with FOLFOX regimen as first line therapy of metastatic colorectal cancer: a feasibility study. Oncology. 78(3-4) 205 - 209 2010. PMID: 20424491. DOI: 10.1159/000313700
41 Kubota Y Han Q Hozumi C Masaki N Yamamoto J Aoki Y Tsunoda T & 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. PMID: 35489727. DOI: 10.21873/anticanres.15734
42 Kokkinakis DM Hoffman RM Frenkel EP Wick JB Han Q Xu M Tan Y & Schold SC Synergy between methionine stress and chemotherapy in the treatment of brain tumor xenografts in athymic mice. Cancer Res. 61(10) 4017 - 4023 2001. PMID: 11358820.
Pubmed |
43 Kawaguchi K Igarashi K Li S Han Q Tan Y Kiyuna T Miyake K Murakami T Chmielowski B Nelson SD Russell TA Dry SM Li Y Unno M Eilber FC & Hoffman RM Combination treatment with recombinant methioninase enables temozolomide to arrest a BRAF V600E melanoma in a patient-derived orthotopic xenograft (PDOX) mouse model. Oncotarget. 8(49) 85516 - 85525 2017. PMID: 29156737. DOI: 10.18632/oncotarget.20231
44 Durando X Thivat E Farges MC Cellarier E D’Incan M Demidem A Vasson MP Barthomeuf C & Chollet P Optimal methionine-free diet duration for nitrourea treatment: a Phase I clinical trial. Nutr Cancer. 60(1) 23 - 30 2008. PMID: 18444132. DOI: 10.1080/01635580701525877
45 Thivat E Farges MC Bacin F D’Incan M Mouret-Reynier MA Cellarier E Madelmont JC Vasson MP Chollet P & Durando X Phase II trial of the association of a methionine-free diet with cystemustine therapy in melanoma and glioma. Anticancer Res. 29(12) 5235 - 5240 2009. PMID: 20044642.
Pubmed |
46 Sugisawa N Higuchi T Han Q Hozumi C Yamamoto J Tashiro Y Nishino H Kawaguchi K Bouvet M Murata T Unno M & Hoffman RM Oral recombinant methioninase combined with paclitaxel arrests recalcitrant ovarian clear cell carcinoma growth in a patient-derived orthotopic xenograft (PDOX) nude-mouse model. Cancer Chemother Pharmacol. 88(1) 61 - 67 2021. PMID: 33768300. DOI: 10.1007/s00280-021-04261-x
47 Lim HI Sun YU Han Q Yamamoto J & Hoffman RM Efficacy of oral recombinant methioninase and eribulin on a PDOX model of triple-negative breast cancer (TNBC) liver metastasis. In Vivo. 35(5) 2531 - 2534 2021. PMID: 34410939. DOI: 10.21873/invivo.12534
48 Aoki Y Tome Y Wu NF Yamamoto J Hamada K Han Q Bouvet M Nishida K & Hoffman RM Oral-recombinant methioninase converts an osteosarcoma from docetaxel-resistant to -sensitive in a clinically-relevant patient-derived orthotopic-xenograft (PDOX) mouse model. Anticancer Res. 41(4) 1745 - 1751 2021. PMID: 33813378. DOI: 10.21873/anticanres.14939
49 Kavya D & Nadumane VK A combination of semi-purified Lmethioninase with tamoxifen citrate to ameliorate breast cancer in athymic nude mice. Mol Biol Rep. PMID: 36566301. DOI: 10.1007/s11033-022-08144-z
50 Igarashi K Kawaguchi K Kiyuna T Miyake K Miyaki M Yamamoto N Hayashi K Kimura H Miwa S Higuchi T Singh AS Chmielowski B Nelson SD Russell TA Eckardt MA Dry SM Li Y Singh SR Chawla SP Eilber FC Tsuchiya H & Hoffman RM Metabolic targeting with recombinant methioninase combined with palbociclib regresses a doxorubicin-resistant dedifferentiated liposarcoma. Biochem Biophys Res Commun. 506(4) 912 - 917 2018. PMID: 30392912. DOI: 10.1016/j.bbrc.2018.10.119
51 Higuchi H Han Q Miyake K Oshiro H Sugisawa N Tan Y Yamamoto N Hayashi K Kimura H Miwa S Igarashi K Bouvet M Singh SR Tsuchiya H & Hoffman RM Combination of oral recombinant methioninase and decitabine arrests a chemotherapy-resistant undifferentiated soft-tissue sarcoma patient-derived orthotopic xenograft mouse model. Biochem Biophys Res Commun. 523 135 - 139 2020. PMID: 31839218. DOI: 10.1016/j.bbrc.2019.12.024.
52 Coalson DW Mecham JO Stern PH & 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. PMID: 6289297. DOI: 10.1073/pnas.79.14.4248
53 Higuchi T Sugisawa N Yamamoto J Oshiro H Han Q Yamamoto N Hayashi K Kimura H Miwa S Igarashi K Tan Y Kuchipudi S Bouvet M Singh SR Tsuchiya H & Hoffman RM The combination of oral-recombinant methioninase and azacitidine arrests achemotherapy-resistant osteosarcoma patient-derived orthotopic xenograft mouse model. Cancer Chemother Pharmacol. 85(2) 285 - 291 2020. PMID: 31705268. DOI: 10.1007/s00280-019-03986-0
54 Sugisawa N Yamamoto J Han Q Tan Y Tashiro Y Nishino H Inubushi S Hamada K Kawaguchi K Unno M Bouvet M & Hoffman RM Triple-methyl blockade with recombinant methioninase, cycloleucine, and azacitidine arrests a pancreatic cancer patient-derived orthotopic xenograft model. Pancreas. 50(1) 93 - 98 2021. PMID: 33370029. DOI: 10.1097/MPA.0000000000001709
55 Marone R Cmiljanovic V Giese B & Wymann MP Targeting phosphoinositide 3-kinase: moving towards therapy. Biochim Biophys Acta. 1784(1) 159 - 185 2008. PMID: 17997386. DOI: 10.1016/j.bbapap.2007.10.003
56 Masaki N Han Q Samonte C Wu NF Hozumi C Wu J Obara K Kubota Y Aoki Y Bouvet M & Hoffman RM Oral-recombinant methioninase in combination with rapamycin eradicates osteosarcoma of the breast in a patient-derived orthotopic xenograft mouse model. Anticancer Res. 42(11) 5217 - 5222 2022. PMID: 36288875. DOI: 10.21873/anticanres.16028
57 Strekalova E Malin D Good DM & Cryns VL Methionine deprivation induces a targetable vulnerability in triple-negative breast cancer cells by enhancing TRAIL Receptor-2 expression. Clin Cancer Res. 21(12) 2780 - 2791 2015. PMID: 25724522. DOI: 10.1158/1078-0432.CCR-14-2792
58 Yamamoto J Miyake K Han Q Tan Y Inubushi S Sugisawa N Higuchi T Tashiro Y Nishino H Homma Y Matsuyama R Chawla SP Bouvet M Singh SR Endo I & Hoffman RM Oral recombinant methioninase increases TRAIL receptor-2 expression to regress pancreatic cancer in combination with agonist tigatuzumab in an orthotopic mouse model. Cancer Lett. 492 174 - 184 2020. PMID: 32739322. DOI: 10.1016/j.canlet.2020.07.034
59 Stern PH Wallace CD & 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. PMID: 6707100. DOI: 10.1002/jcp.1041190106
60 Tan Y Xu M & 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. PMID: 20530407.
Pubmed |
61 Yano S Li S Han Q Tan Y Bouvet M Fujiwara T & 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. PMID: 25238266. DOI: 10.18632/oncotarget.2369
62 Aoki Y Tome Y Han Q Yamamoto J Hamada K Masaki N Bouvet M Nishida K & 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. PMID: 34877414. DOI: 10.1016/j.bbrep.2021.101177
63 Hoffman RM Coalson DW Jacobsen SJ & Erbe RW Folate polyglutamate and monoglutamate accumulation in normal and SV40-transformed human fibroblasts. J Cell Physiol. 109(3) 497 - 505 1981. PMID: 6274882. DOI: 10.1002/jcp.1041090316
64 Ghergurovich JM Xu X Wang JZ Yang L Ryseck RP Wang L & Rabinowitz JD Methionine synthase supports tumour tetrahydrofolate pools. Nat Metab. 3(11) 1512 - 1520 2021. PMID: 34799699. DOI: 10.1038/s42255-021-00465-w
65 Sullivan MR Darnell AM Reilly MF Kunchok T Joesch-Cohen L Rosenberg D Ali A Rees MG Roth JA Lewis CA & Vander Heiden MG Methionine synthase is essential for cancer cell proliferation in physiological folate environments. Nat Metab. 3(11) 1500 - 1511 2021. PMID: 34799701. DOI: 10.1038/s42255-021-00486-5
66 Halpern BC Clark BR Hardy DN Halpern RM & Smith RA The effect of replacement of methionine by homocystine on survival of malignant and normal adult mammalian cells in culture. Proc Natl Acad Sci USA. 71(4) 1133 - 1136 1974. PMID: 4524624. DOI: 10.1073/pnas.71.4.1133
67 Ashe H Clark BR Chu F Hardy DN Halpern BC Halpern RM & Smith RA N5-methyltetrahydrofolate: homocysteine methyltransferase activity in extracts from normal, malignant and embryonic tissue culture cells. Biochem Biophys Res Commun. 57(2) 417 - 425 1974. PMID: 452450. DOI: 10.1016/0006-291x(74)90947-4
68 Sorin M Watkins D Gilfix BM & Rosenblatt DS Methionine dependence in tumor cells: The potential role of cobalamin and MMACHC. Mol Genet Metab. 132(3) 155 - 161 2021. PMID: 33487542. DOI: 10.1016/j.ymgme.2021.01.006
69 Park M Leahey E & Funk RJ Papers and patents are becoming less disruptive over time. Nature. 613(7942) 138 - 144 2023. PMID: 36600070. DOI: 10.1038/s41586-022-05543-x
70 Kaiser P Methionine dependence of cancer. Biomolecules. 10(4) 568 2020. PMID: 32276408. DOI: 10.3390/biom10040568
71 Montalbano S Raboni S Sidoli S Mozzarelli A Bettati S & 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. PMID: 36672476. DOI: 10.3390/cancers15020527
72 Jacobsen SJ Hoffman RM & 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.
Pubmed |
73 Mecham JO Rowitch D Wallace CD Stern PH & Hoffman RM The metabolic defect of methionine dependence occurs frequently in human tumor cell lines. Biochem Biophys Res Commun. 117(2) 429 - 434 1983. PMID: 6661235. DOI: 10.1016/0006-291x(83)91218-4
74 Malin D Lee Y Chepikova O Strekalova E Carlson A & Cryns VL Methionine restriction exposes a targetable redox vulnerability of triple-negative breast cancer cells by inducing thioredoxin reductase. Breast Cancer Res Treat. 190(3) 373 - 387 2021. PMID: 34553295. DOI: 10.1007/s10549-021-06398-y
cdp > Vol 3 - 3 > Pages 272 - 281