Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
In Vivo
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
In Vivo

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Research ArticleClinical Studies
Open Access

Extensive Shrinkage and Long-term Stable Disease in a Teenage Female Patient With High-grade Glioma Treated With Temozolomide and Radiation in Combination With Oral Recombinant Methioninase and a Low-methionine Diet

MOTOKAZU SATO, QINGHONG HAN, KOHEI MIZUTA, RYOSUKE MORI, BYUNG MO KANG, SEI MORINAGA, NORITOSHI KOBAYASHI, YASUSHI ICHIKAWA, ATSUSHI NAKAJIMA and ROBERT M. HOFFMAN
In Vivo May 2024, 38 (3) 1459-1464; DOI: https://doi.org/10.21873/invivo.13591
MOTOKAZU SATO
1AntiCancer Inc., San Diego, CA, U.S.A.;
2Department of Surgery, University of California, San Diego, CA, U.S.A.;
3Department of Oncology, Yokohama City University School of Medicine, Yokohama, Japan;
4Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
QINGHONG HAN
1AntiCancer Inc., San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
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
RYOSUKE MORI
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
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
SEI MORINAGA
1AntiCancer Inc., San Diego, CA, U.S.A.;
2Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
NORITOSHI KOBAYASHI
3Department of Oncology, Yokohama City University School of Medicine, Yokohama, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YASUSHI ICHIKAWA
3Department of Oncology, Yokohama City University School of Medicine, Yokohama, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ATSUSHI NAKAJIMA
4Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ROBERT M. HOFFMAN
1AntiCancer Inc., San Diego, CA, U.S.A.;
2Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: all@anticancer.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Gliomas are the most common and recalcitrant malignant primary brain tumors. All cancer types are addicted to methionine, which is a fundamental and general hallmark of cancer known as the Hoffman effect. Particularly glioma cells exhibit methionine addiction. Because of methionine addiction, [11C]-methionine positron emission tomography (MET-PET) is widely used for glioma imaging in clinical practice, which can monitor the extent of methionine addiction. Methionine restriction including recombinant methioninase (rMETase) and a low-methionine diet, has shown high efficacy in preclinical models of gliomas, especially in combination with chemotherapy. The aim of the present study was to determine the efficacy of methionine restriction with oral rMETase (o-rMETase) and a low-methionine diet, combined with radiation and temozolomide (TMZ), on a teenage female patient with high-grade glioma. Case Report: A 16-year-old girl was diagnosed with high-grade glioma. Magnetic resonance imaging (MRI) showed a left temporal-lobe tumor with compression to the left lateral ventricle and narrowing of sulci in the left temporal lobe. After the start of methionine restriction with o-rMETase and a low-methionine diet, along with TMZ combined with radiotherapy, the tumor size shrunk at least 60%, with improvement in the left lateral ventricle and sulci. The patient’s condition remains stable for 19 months without severe adverse effects. Conclusion: Methionine restriction consisting of o-rMETase and a low-methionine diet, in combination with radiation and TMZ as first-line chemotherapy, were highly effective in a patient with high-grade glioma.

Key Words:
  • High-grade glioma
  • temozolomide
  • radiation
  • methionine addiction
  • Hoffman effect
  • methionine restriction
  • oral methioninase
  • low-methionine diet
  • combination therapy
  • synergy

Gliomas are the most common and recalcitrant primary brain and other nervous-system malignant tumors in the United States (1). Although multidisciplinary therapies such as surgical resection, radiation and chemotherapy have been introduced, gliomas are prone to recurrence and have a very poor prognosis (2). Especially for high grade glioma, the 5-year survival rate is 6.7% (1) and the average survival is 9 months (3).

All cancer types are addicted to methionine, which is a fundamental and general hallmark of cancer known as the Hoffman effect (4-7). Methionine addiction is at least partly due to excess transmethylation in cancer cells, leading to a much higher demand for external methionine in comparison to normal cells (4, 8-12). Therefore, cancer cells are incapable of surviving without exogenous methionine, despite their high endogenous production of methionine (4, 10-12).

Glioma cells are particularly methionine addicted. [11C]-methionine positron emission tomography (MET-PET) is used for diagnosis (13, 14), treatment (15, 16) and to estimate prognosis (17, 18) in glioma. MET-PET yields a stronger and more accurate signal than [18F]-deoxyglucose PET (FDG-PET), as gliomas are more methionine addicted than glucose addicted (19). Moreover, a reduced intake of methionine seen on MET-PET is linked to a better prognosis of malignant glioma (20, 21).

Temozolomide (TMZ) is an oral alkylating agent used as first-line chemotherapy for glioma (22). TMZ functions by methylating DNA resulting in DNA damage, causing cell-cycle arrest in the G2/M phase and death of glioma cells (23). However, the improvement of overall survival by TMZ is limited (24).

Our laboratory has developed recombinant methioninase (rMETase) for methionine restriction (25). When administered orally (o-rMETase), this enzyme breaks down methionine in the gut, leading to a reduction in methionine levels in the circulation and in tumors (26). Since, methionine restriction, including rMETase, selectively arrests the cell cycle of cancer cells in the late-S/G2 phase (27, 28), alkylating drugs showed a synergistic effect when combined with methionine restriction in mouse models of glioblastoma, since they are methionine addicted (29-35).

In the present case report, a 16-year-old girl with high-grade glioma treated with radiation and temozolomide combined with oral rMETase (o-rMETase) and a low-methionine diet, demonstrated extensive tumor shrinkage and subsequent stable disease for 19 months, thus far.

Materials and Methods

Production and formulation of rMETase. rMETase was produced by fermenting recombinant Escherichia coli transformed with the methioninase gene from Pseudomonas putida. The purification of methioninase involved a high-yield method that had a heat step at 60°C, polyethylene glycol precipitation, and ion-exchange column chromatography using diethylaminoethyl (DEAE)-Sepharose FF (36, 37).

Methionine restriction and o-rMETase administration. The patient stayed on a methionine restricted diet, following the Nutritional Oncology Research Institute (NORI) protocol (38). This procedure recommends less than 2 mg/kg body weight methionine intake per day. o-rMETase was administered orally twice a day 30 min after each meal at a dose of 250 units, as a supplement.

Case Report

A 16-year-old girl was diagnosed with high-grade glioma in July 2022. Magnetic resonance imaging (MRI) showed a left temporal lobe tumor with compression to the left lateral ventricle and narrowing of sulci in the left temporal lobe (Figure 1A).

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

MRI images of the brain of a 16-year-old girl with high grade glioma. A) The tumor was located in the left temporal lobe with compression of the left lateral ventricle and narrowing of left temporal lobe sulci before the start of treatment in July 2022. The patient began methionine restriction with o-rMETase and a low-methionine diet along with TMZ (75 mg/m2) combined with radiotherapy in August 2022. After completion of radiotherapy, the dose of TMZ was increased to 150 mg/m3. B) The tumor shrunk with improvement of the left lateral ventricle and left temporal lobe sulci in March 2023. o-rMETase: Oral recombinant methioninase; TMZ: temozolomide.

The patient began methionine restriction with o-rMETase and a low-methionine diet along with TMZ (75 mg/m2) combined with radiotherapy in August 2022. After she completed radiotherapy in October 2022, the dose of TMZ was increased to 150 mg/m2.

An MRI showed an extensive reduction in the size of the tumor, along with improvement in the lateral ventricles and sulci by nine months after the start of treatment (Figure 1B). The tumor shrunk at least 60% by nine months of treatment with radiation and TMZ combined with o-rMETase and a low-methionine diet and is currently stable after 19 months (Figure 2). This allows her to maintain the current combination treatment of TMZ and o-rMETase and a low-methionine diet without severe adverse effects.

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

Relative-brain tumor volume over time of treatment. The patient began methionine restriction with o-rMETase, and a low-methionine diet along with TMZ (75 mg/m2) combined with radiotherapy in August 2022. After completion of radiotherapy, the dose of TMZ was increased to 150 mg/m3. The tumor volume at diagnosis in July 2022 was designated as 1. o-rMETase: Oral recombinant methioninase; TMZ: temozolomide.

Discussion

This is the first clinical report of combining TMZ and methionine restriction, comprising oral rMETase and a low-methionine diet, on a patient with high-grade glioma.

In a clinical trial, the median survival was 14.6 months among high-grade glioma patients treated with radiation and TMZ (24). However, patients who participate in clinical trials usually have better performance status (PS) and often more stable disease than the general population of high-grade glioma. The median survival in the general population of high-grade glioma was reported to be 9 months (3). It is noteworthy that the patient in the present study has maintained a good PS for 19 months since diagnosis.

Since gliomas are heterogeneous and resistant to all modalities of standard treatment including surgery, radiation and TMZ, which has not changed for 20 years despite many attempts at implementing new treatments (39). An effective treatment is urgently required.

Methionine addiction is a fundamental and general hallmark of all cancer types, including glioma, as seen in preclinical studies (29-35). Methionine restriction with rMETase and a low-methionine diet greatly enhanced the efficacy of an alkylating agent in mouse models of brain cancer (31, 32, 35).

Histone-methylation alteration in cancer is linked to methionine addiction (8, 40, 47). S-adenosylmethionine (SAM), the main metabolite of MET and universal methyl donor in the cell, contributes to the malignancy of cancer (10, 40). Suppressing SAM by methionine restriction inhibits glioma growth (41).

We first found that methionine restriction is synergistic with chemotherapy almost 40 years ago (42). Subsequently, a large amount of research has shown that there is synergy between methionine restriction, specifically with rMETase and/or a low-methionine diet, and all types of chemotherapy (43).

The present study suggests that methionine restriction is promising in combination with radiation and chemotherapy for high grade glioma. Methionine restriction does not involve the blood-brain barrier. Further clinical studies are necessary.

o-rMETase is effective as it targets the fundamental basis of cancer (4-12, 27, 28, 40, 44-57) and has shown potential efficacy in the clinic for other cancers including prostate cancer (58-60), colorectal cancer (61, 62), pancreatic cancer (63) and breast cancer (64).

Recent studies have suggested that MAT2A and histone methylation are therapeutic targets for diffuse midline gliomas and other cancers (65, 66). The present and previous results indicate that methionine addiction is a superior and safer target for gliomas and other cancers (4-10, 29-35, 40, 46-54).

Acknowledgements

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

Footnotes

  • Authors’ Contributions

    MS and RMH wrote the article. MS, QH, RM, KM, SM, BMK, NK, YI, AN, and RMH reviewed the article.

  • Conflicts of Interest

    The Authors declare no competing interests regarding this work.

  • Funding

    The Robert M. Hoffman Foundation for Cancer Research provided funds for this study.

  • Received February 2, 2024.
  • Revision received March 4, 2024.
  • Accepted March 5, 2024.
  • Copyright © 2024, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Ostrom QT,
    2. Price M,
    3. Neff C,
    4. Cioffi G,
    5. Waite KA,
    6. Kruchko C,
    7. Barnholtz-Sloan JS
    : CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2016-2020. Neuro Oncol 25(12 Suppl 2): iv1-iv99, 2023. DOI: 10.1093/neuonc/noad149
    OpenUrlCrossRef
  2. ↵
    1. Miller KD,
    2. Ostrom QT,
    3. Kruchko C,
    4. Patil N,
    5. Tihan T,
    6. Cioffi G,
    7. Fuchs HE,
    8. Waite KA,
    9. Jemal A,
    10. Siegel RL,
    11. Barnholtz-Sloan JS
    : Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin 71: 381-406, 2021. DOI: 10.3322/caac.21693
    OpenUrlCrossRefPubMed
  3. ↵
    1. Brown NF,
    2. Ottaviani D,
    3. Tazare J,
    4. Gregson J,
    5. Kitchen N,
    6. Brandner S,
    7. Fersht N,
    8. Mulholland P
    : Survival outcomes and prognostic factors in glioblastoma. Cancers (Basel) 14(13): 3161, 2022. DOI: 10.3390/cancers14133161
    OpenUrlCrossRef
  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
    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
  5. ↵
    1. Kaiser P
    : Methionine dependence of cancer. Biomolecules 10(4): 568, 2020. DOI: 10.3390/biom10040568
    OpenUrlCrossRefPubMed
  6. ↵
    1. Yamamoto J,
    2. Han Q,
    3. Inubushi S,
    4. Sugisawa N,
    5. Hamada K,
    6. Nishino H,
    7. Miyake K,
    8. Kumamoto T,
    9. Matsuyama R,
    10. Bouvet M,
    11. Endo I,
    12. Hoffman RM
    : Histone methylation status of H3K4me3 and H3K9me3 under methionine restriction is unstable in methionine-addicted cancer cells, but stable in normal cells. Biochem Biophys Res Commun 533(4): 1034-1038, 2020. DOI: 10.1016/j.bbrc.2020.09.108
    OpenUrlCrossRefPubMed
    1. 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. 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. 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
  8. ↵
    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
  9. ↵
    1. Bag AK,
    2. Wing MN,
    3. Sabin ND,
    4. Hwang SN,
    5. Armstrong GT,
    6. Han Y,
    7. Li Y,
    8. Snyder SE,
    9. Robinson GW,
    10. Qaddoumi I,
    11. Broniscer A,
    12. Lucas JT,
    13. Shulkin BL
    : (11)C-Methionine PET for identification of pediatric high-grade glioma recurrence. J Nucl Med 63(5): 664-671, 2022. DOI: 10.2967/jnumed.120.261891
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Galldiks N,
    2. Lohmann P,
    3. Langen KJ
    : The role of 11C-methionine PET in patients with newly diagnosed WHO grade 2 or 3 gliomas. Neuro Oncol 24(9): 1557-1558, 2022. DOI: 10.1093/neuonc/noac120
    OpenUrlCrossRef
  11. ↵
    1. Jacobo JA,
    2. Buentello M,
    3. Del Valle R
    : C-methionine-PET-guided Gamma Knife radiosurgery boost as adjuvant treatment for newly diagnosed glioblastomas. Surg Neurol Int 12: 247, 2021. DOI: 10.25259/SNI_706_2020
    OpenUrlCrossRef
  12. ↵
    1. Ohmura K,
    2. Daimon T,
    3. Ikegame Y,
    4. Yano H,
    5. Yokoyama K,
    6. Kumagai M,
    7. Shinoda J,
    8. Iwama T
    : Resection of positive tissue on methionine-PET is associated with improved survival in glioblastomas. Brain Behav 13(12): e3291, 2023. DOI: 10.1002/brb3.3291
    OpenUrlCrossRef
  13. ↵
    1. Jung TY,
    2. Min JJ,
    3. Bom HS,
    4. Jung S,
    5. Kim IY,
    6. Lim SH,
    7. Kim DY,
    8. Kwon SY
    : Prognostic value of post-treatment metabolic tumor volume from 11C-methionine PET/CT in recurrent malignant glioma. Neurosurg Rev 40(2): 223-229, 2017. DOI: 10.1007/s10143-016-0748-1
    OpenUrlCrossRefPubMed
  14. ↵
    1. Nakajo K,
    2. Uda T,
    3. Kawashima T,
    4. Terakawa Y,
    5. Ishibashi K,
    6. Tsuyuguchi N,
    7. Tanoue Y,
    8. Nagahama A,
    9. Uda H,
    10. Koh S,
    11. Sasaki T,
    12. Ohata K,
    13. Kanemura Y,
    14. Goto T
    : Maximum 11C-methionine PET uptake as a prognostic imaging biomarker for newly diagnosed and untreated astrocytic glioma. Sci Rep 12(1): 546, 2022. DOI: 10.1038/s41598-021-04216-5
    OpenUrlCrossRef
  15. ↵
    1. Kubota Y,
    2. Sato T,
    3. Hozumi C,
    4. Han Q,
    5. Aoki Y,
    6. Masaki N,
    7. Obara K,
    8. Tsunoda T,
    9. Hoffman RM
    : Superiority of [(11)C]methionine over [(18)F]deoxyglucose for PET imaging of multiple cancer types due to the methionine addiction of cancer. Int J Mol Sci 24(3): 1935, 2023. DOI: 10.3390/ijms24031935
    OpenUrlCrossRefPubMed
  16. ↵
    1. Kato T,
    2. Shinoda J,
    3. Oka N,
    4. Miwa K,
    5. Nakayama N,
    6. Yano H,
    7. Maruyama T,
    8. Muragaki Y,
    9. Iwama T
    : Analysis of 11C-methionine uptake in low-grade gliomas and correlation with proliferative activity. AJNR Am J Neuroradiol 29(10): 1867-1871, 2008. DOI: 10.3174/ajnr.A1242
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Kobayashi K,
    2. Hirata K,
    3. Yamaguchi S,
    4. Manabe O,
    5. Terasaka S,
    6. Kobayashi H,
    7. Shiga T,
    8. Hattori N,
    9. Tanaka S,
    10. Kuge Y,
    11. Tamaki N
    : Prognostic value of volume-based measurements on 11C-methionine PET in glioma patients. Eur J Nucl Med Mol Imaging 42(7): 1071-1080, 2015. DOI: 10.1007/s00259-015-3046-1
    OpenUrlCrossRefPubMed
  18. ↵
    1. Schaff LR,
    2. Mellinghoff IK
    : Glioblastoma and other primary brain malignancies in adults. JAMA 329(7): 574, 2023. DOI: 10.1001/jama.2023.0023
    OpenUrlCrossRef
  19. ↵
    1. Tomar MS,
    2. Kumar A,
    3. Srivastava C,
    4. Shrivastava A
    : Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochim Biophys Acta Rev Cancer 1876(2): 188616, 2021. DOI: 10.1016/j.bbcan.2021.188616
    OpenUrlCrossRefPubMed
  20. ↵
    1. Stupp R,
    2. Mason WP,
    3. van den Bent MJ,
    4. Weller M,
    5. Fisher B,
    6. Taphoorn MJ,
    7. Belanger K,
    8. Brandes AA,
    9. Marosi C,
    10. Bogdahn U,
    11. Curschmann J,
    12. Janzer RC,
    13. Ludwin SK,
    14. Gorlia T,
    15. Allgeier A,
    16. Lacombe D,
    17. Cairncross JG,
    18. Eisenhauer E,
    19. Mirimanoff RO, European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups, National Cancer Institute of Canada Clinical Trials Group
    : Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10): 987-996, 2005. DOI: 10.1056/NEJMoa043330
    OpenUrlCrossRefPubMed
  21. ↵
    1. Hoffman RM
    : Development of recombinant methioninase to target the general cancer-specific metabolic defect of methionine dependence: a 40-year odyssey. Expert Opin Biol Ther 15(1): 21-31, 2015. DOI: 10.1517/14712598.2015.963050
    OpenUrlCrossRefPubMed
  22. ↵
    1. Kawaguchi K,
    2. Han Q,
    3. Li S,
    4. Tan Y,
    5. Igarashi K,
    6. Kiyuna T,
    7. Miyake K,
    8. Miyake M,
    9. Chmielowski B,
    10. Nelson SD,
    11. Russell TA,
    12. Dry SM,
    13. Li Y,
    14. Singh AS,
    15. Eckardt MA,
    16. Unno M,
    17. Eilber FC,
    18. Hoffman RM
    : Targeting methionine with oral recombinant methioninase (o-rMETase) arrests a patient-derived orthotopic xenograft (PDOX) model of BRAF-V600E mutant melanoma: implications for chronic clinical cancer therapy and prevention. Cell Cycle 17(3): 356-361, 2018. DOI: 10.1080/15384101.2017.1405195
    OpenUrlCrossRefPubMed
  23. ↵
    1. Hoffman RM,
    2. Jacobsen SJ
    : Reversible growth arrest in simian virus 40-transformed human fibroblasts. Proc Natl Acad Sci U S A 77(12): 7306-7310, 1980. DOI: 10.1073/pnas.77.12.7306
    OpenUrlAbstract/FREE Full Text
  24. ↵
    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
  25. ↵
    1. Poirson-Bichat F,
    2. Lopez R,
    3. Bras Gonçalves RA,
    4. Miccoli L,
    5. Bourgeois Y,
    6. Demerseman P,
    7. Poisson M,
    8. Dutrillaux B,
    9. Poupon MF
    : Methionine deprivation and methionine analogs inhibit cell proliferation and growth of human xenografted gliomas. Life Sci 60(12): 919-931, 1997. DOI: 10.1016/s0024-3205(96)00672-8
    OpenUrlCrossRefPubMed
    1. Poirson-Bichat F,
    2. Gonçalves RA,
    3. Miccoli L,
    4. Dutrillaux B,
    5. Poupon MF
    : Methionine depletion enhances the antitumoral efficacy of cytotoxic agents in drug-resistant human tumor xenografts. Clin Cancer Res 6: 643-653, 2000.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Kokkinakis DM,
    2. Hoffman RM,
    3. Frenkel EP,
    4. Wick JB,
    5. Han Q,
    6. Xu M,
    7. Tan Y,
    8. Schold SC
    : Synergy between methionine stress and chemotherapy in the treatment of brain tumor xenografts in athymic mice. Cancer Res 61: 4017-4023, 2001.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Kokkinakis DM,
    2. Wick JB,
    3. Zhou QX
    : Metabolic response of normal and malignant tissue to acute and chronic methionine stress in athymic mice bearing human glial tumor xenografts. Chem Res Toxicol 15(11): 1472-1479, 2002. DOI: 10.1021/tx020033n
    OpenUrlCrossRefPubMed
    1. Liu H,
    2. Zhang W,
    3. Wang K,
    4. Wang X,
    5. Yin F,
    6. Li C,
    7. Wang C,
    8. Zhao B,
    9. Zhong C,
    10. Zhang J,
    11. Peng F,
    12. Bi Y,
    13. Shen C,
    14. Hou X,
    15. Zhang D,
    16. Liu Y,
    17. Ai J,
    18. Zhao S
    : Methionine and cystine double deprivation stress suppresses glioma proliferation via inducing ROS/autophagy. Toxicol Lett 232(2): 349-355, 2015. DOI: 10.1016/j.toxlet.2014.11.011
    OpenUrlCrossRef
    1. Upadhyayula PS,
    2. Higgins DM,
    3. Mela A,
    4. Banu M,
    5. Dovas A,
    6. Zandkarimi F,
    7. Patel P,
    8. Mahajan A,
    9. Humala N,
    10. Nguyen TTT,
    11. Chaudhary KR,
    12. Liao L,
    13. Argenziano M,
    14. Sudhakar T,
    15. Sperring CP,
    16. Shapiro BL,
    17. Ahmed ER,
    18. Kinslow C,
    19. Ye LF,
    20. Siegelin MD,
    21. Cheng S,
    22. Soni R,
    23. Bruce JN,
    24. Stockwell BR,
    25. Canoll P
    : Dietary restriction of cysteine and methionine sensitizes gliomas to ferroptosis and induces alterations in energetic metabolism. Nat Commun 14(1): 1187, 2023. DOI: 10.1038/s41467-023-36630-w
    OpenUrlCrossRef
  28. ↵
    1. Kubota Y,
    2. Aoki Y,
    3. Masaki N,
    4. Obara K,
    5. Hamada K,
    6. Han Q,
    7. Bouvet M,
    8. Tsunoda T,
    9. Hoffman RM
    : Methionine restriction of glioma does not induce MGMT and greatly improves temozolomide efficacy in an orthotopic nude-mouse model: A potential curable approach to a clinically-incurable disease. Biochem Biophys Res Commun 695: 149418, 2024. DOI: 10.1016/j.bbrc.2023.149418
    OpenUrlCrossRef
  29. ↵
    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
  30. ↵
    1. Takakura T,
    2. Ito T,
    3. Yagi S,
    4. Notsu Y,
    5. Itakura T,
    6. Nakamura T,
    7. Inagaki K,
    8. Esaki N,
    9. Hoffman RM,
    10. Takimoto A
    : High-level expression and bulk crystallization of recombinant l-methionine γ-lyase, an anticancer agent. Appl Microbiol Biotechnol 70(2): 183-192, 2006. DOI: 10.1007/s00253-005-0038-2
    OpenUrlCrossRefPubMed
  31. ↵
    How to Starve Cancer Naturally. Starve Cancer Nat. Available at: https://howtostarvecancernaturally.com/methionine-chart [Last accessed on December 22, 2023]
  32. ↵
    1. Wen PY,
    2. Weller M,
    3. Lee EQ,
    4. Alexander BM,
    5. Barnholtz-Sloan JS,
    6. Barthel FP,
    7. Batchelor TT,
    8. Bindra RS,
    9. Chang SM,
    10. Chiocca EA,
    11. Cloughesy TF,
    12. DeGroot JF,
    13. Galanis E,
    14. Gilbert MR,
    15. Hegi ME,
    16. Horbinski C,
    17. Huang RY,
    18. Lassman AB,
    19. Le Rhun E,
    20. Lim M,
    21. Mehta MP,
    22. Mellinghoff IK,
    23. Minniti G,
    24. Nathanson D,
    25. Platten M,
    26. Preusser M,
    27. Roth P,
    28. Sanson M,
    29. Schiff D,
    30. Short SC,
    31. Taphoorn MJB,
    32. Tonn JC,
    33. Tsang J,
    34. Verhaak RGW,
    35. von Deimling A,
    36. Wick W,
    37. Zadeh G,
    38. Reardon DA,
    39. Aldape KD,
    40. van den Bent MJ
    : Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol 22(8): 1073-1113, 2020. DOI: 10.1093/neuonc/noaa106
    OpenUrlCrossRefPubMed
  33. ↵
    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
  34. ↵
    1. Harachi M,
    2. Masui K,
    3. Honda H,
    4. Muragaki Y,
    5. Kawamata T,
    6. Cavenee WK,
    7. Mischel PS,
    8. Shibata N
    : Dual regulation of histone methylation by mTOR complexes controls glioblastoma tumor cell growth via EZH2 and SAM. Mol Cancer Res 18(8): 1142-1152, 2020. DOI: 10.1158/1541-7786.MCR-20-0024
    OpenUrlAbstract/FREE Full Text
  35. ↵
    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
  36. ↵
    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
    OpenUrlCrossRef
  37. ↵
    1. Kubota Y,
    2. Sato T,
    3. Han Q,
    4. Hozumi C,
    5. Morinaga S,
    6. Mizuta K,
    7. Tsunoda T,
    8. Hoffman RM
    : [11C] Methionine-PET imaging as a cancer biomarker for methionine addiction and sensitivity to methionine-restriction-based combination chemotherapy. In Vivo 38(1): 253-258, 2024. DOI: 10.21873/invivo.13432
    OpenUrlAbstract/FREE Full Text
    1. Pokrovsky VS,
    2. Qoura LA,
    3. Demidova EA,
    4. Han Q,
    5. Hoffman RM
    : Targeting methionine addiction of cancer cells with methioninase. Biochemistry (Mosc) 88(7): 944-952, 2023. DOI: 10.1134/S0006297923070076
    OpenUrlCrossRef
  38. ↵
    1. Aoki Y,
    2. Han Q,
    3. Kubota Y,
    4. Masaki N,
    5. Obara K,
    6. Tome Y,
    7. Bouvet M,
    8. Nishida K,
    9. Hoffman RM
    : Oncogenes and methionine addiction of cancer: Role of c-MYC. Cancer Genomics Proteomics 20(2): 165-170, 2023. DOI: 10.21873/cgp.20371
    OpenUrlAbstract/FREE Full Text
  39. ↵
    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. Aoki Y,
    2. Tome Y,
    3. Han Q,
    4. Yamamoto J,
    5. Hamada K,
    6. Masaki N,
    7. Kubota Y,
    8. Bouvet M,
    9. Nishida K,
    10. Hoffman RM
    : Deletion of MTAP highly sensitizes osteosarcoma cells to methionine restriction with recombinant methioninase. Cancer Genomics Proteomics 19(3): 299-304, 2022. DOI: 10.21873/cgp.20321
    OpenUrlAbstract/FREE Full Text
    1. 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. Aoki Y,
    2. Tome Y,
    3. Han Q,
    4. Yamamoto J,
    5. Hamada K,
    6. Masaki N,
    7. Bouvet M,
    8. Nishida K,
    9. Hoffman RM
    : Histone H3 lysinetrimethylation markers are decreased by recombinant methioninase and increased by methotrexate at concentrations which inhibit methionine-addicted osteosarcoma cell proliferation. Biochem Biophys Rep 28:101177, 2021. DOI: 10.1016/j.bbrep.2021.101177
    OpenUrlCrossRefPubMed
    1. Aoki Y,
    2. Yamamoto J,
    3. Tome Y,
    4. Hamada K,
    5. Masaki N,
    6. Inubushi S,
    7. Tashiro Y,
    8. Bouvet M,
    9. Endo I,
    10. Nishida K,
    11. Hoffman RM
    : Overmethylation of Histone H3 Lysines is a common molecular change among the three major types of soft-tissue sarcoma in patientderived xenograft (PDX) mouse models. Cancer Genomics Proteomics 18(6): 715-721, 2021. DOI: 10.21873/cgp.20292
    OpenUrlAbstract/FREE Full Text
    1. Yamamoto J,
    2. Aoki Y,
    3. Han Q,
    4. Sugisawa N,
    5. Sun YU,
    6. Hamada K,
    7. Nishino H,
    8. Inubushi S,
    9. Miyake K,
    10. Matsuyama R,
    11. Bouvet M,
    12. Endo I,
    13. Hoffman RM
    : Reversion from methionine addiction to methionine independence results in loss of tumorigenic potential of highly-malignant lung-cancer cells. Anticancer Res 41(2): 641-643, 2021. DOI: 10.21873/anticanres.14815
    OpenUrlAbstract/FREE Full Text
    1. Hoffman RM,
    2. Jacobsen SJ,
    3. Erbe RW
    : Reversion to methionine independence in simian virus 40-transformed human and malignant rat fibroblasts is associated with altered ploidy and altered properties of transformation. Proc Natl Acad Sci 76(3): 1313-1317, 1979. DOI: 10.1073/pnas.76.3.1313
    OpenUrlAbstract/FREE Full Text
  40. ↵
    1. Hoffman RM,
    2. Jacobsen SJ,
    3. Erbe RW
    : Reversion to methionine independence by malignant rat and SV40-transformed human fibroblasts. Biochem Biophys Res Commun 82(1): 228-234, 1978. DOI: 10.1016/0006-291x(78)90600-9
    OpenUrlCrossRefPubMed
    1. Mecham JO,
    2. Rowitch D,
    3. Wallace CD,
    4. Stern PH,
    5. Hoffman RM
    : The metabolic defect of methionine dependence occurs frequently in human tumor cell lines. Biochem Biophys Res Commun 117(2): 429-34, 1983. DOI: 10.1016/0006-291x(83)91218-4
    OpenUrlCrossRefPubMed
    1. Tan Y,
    2. Xu M,
    3. Hoffman RM
    : Broad selective efficacy of recombinant methioninase and polyethylene glycol-modified recombinant methioninase on cancer cells In Vitro. Anticancer Res 30(4): 1041-6, 2010.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    1. Stern PH,
    2. Wallace CD,
    3. Hoffman RM
    : Altered methionine metabolism occurs in all members of a set of diverse human tumor cell lines. J Cell Physiol 119(1): 29-34, 1984. DOI: 10.1002/jcp.104119010658
    OpenUrlCrossRefPubMed
  42. ↵
    1. Han Q,
    2. Tan Y,
    3. Hoffman RM
    : Oral dosing of Recombinant Methioninase Is Associated With a 70% Drop in PSA in a Patient With Bone-metastatic Prostate Cancer and 50% Reduction in Circulating Methionine in a High-stage Ovarian Cancer Patient. Anticancer Res 40(5): 2813-2819, 2020. DOI: 10.21873/anticanres.14254
    OpenUrlAbstract/FREE Full Text
    1. Han Q,
    2. Hoffman RM
    : Chronic Treatment of an Advanced Prostate-cancer Patient With Oral Methioninase Resulted in Long-term Stabilization of Rapidly Rising PSA Levels. In Vivo 35(4): 2171-2176, 2021. DOI: 10.21873/invivo.12488
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Han Q,
    2. Hoffman RM
    : Lowering and Stabilizing PSA Levels in Advanced-prostate Cancer Patients With Oral Methioninase. Anticancer Res 41(4): 1921-1926, 2021. DOI: 10.21873/anticanres.14958
    OpenUrlAbstract/FREE Full Text
  44. ↵
    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 lowmethionine 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
  45. ↵
    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 rectalcancer 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
  46. ↵
    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
  47. ↵
    1. Kubota Y,
    2. Han Q,
    3. Masaki N,
    4. Hozumi C,
    5. Hamada K,
    6. Aoki Y,
    7. Obara K,
    8. Tsunoda T,
    9. Hoffman RM
    : Elimination of axillarylymph-node metastases in a patient with invasive lobular breast cancer treated by first-line neo-adjuvant chemotherapy combined with methionine restriction. Anticancer Res 42(12): 5819-5823, 2022. DOI: 10.21873/anticanres.16089
    OpenUrlAbstract/FREE Full Text
  48. ↵
    1. Golbourn BJ,
    2. Halbert ME,
    3. Halligan K,
    4. Varadharajan S,
    5. Krug B,
    6. Mbah NE,
    7. Kabir N,
    8. Stanton AJ,
    9. Locke AL,
    10. Casillo SM,
    11. Zhao Y,
    12. Sanders LM,
    13. Cheney A,
    14. Mullett SJ,
    15. Chen A,
    16. Wassell M,
    17. Andren A,
    18. Perez J,
    19. Jane EP,
    20. Premkumar DRD,
    21. Koncar RF,
    22. Mirhadi S,
    23. McCarl LH,
    24. Chang YF,
    25. Wu YL,
    26. Gatesman TA,
    27. Cruz AF,
    28. Zapotocky M,
    29. Hu B,
    30. Kohanbash G,
    31. Wang X,
    32. Vartanian A,
    33. Moran MF,
    34. Lieberman F,
    35. Amankulor NM,
    36. Wendell SG,
    37. Vaske OM,
    38. Panigrahy A,
    39. Felker J,
    40. Bertrand KC,
    41. Kleinman CL,
    42. Rich JN,
    43. Friedlander RM,
    44. Broniscer A,
    45. Lyssiotis C,
    46. Jabado N,
    47. Pollack IF,
    48. Mack SC,
    49. Agnihotri S
    : Loss of MAT2A compromises methionine metabolism and represents a vulnerability in H3K27M mutant glioma by modulating the epigenome. Nat Cancer 3(5): 629-648, 2022. DOI: 10.1038/s43018-022-00348-3
    OpenUrlCrossRef
  49. ↵
    1. Yamagishi M,
    2. Kuze Y,
    3. Kobayashi S,
    4. Nakashima M,
    5. Morishima S,
    6. Kawamata T,
    7. Makiyama J,
    8. Suzuki K,
    9. Seki M,
    10. Abe K,
    11. Imamura K,
    12. Watanabe E,
    13. Tsuchiya K,
    14. Yasumatsu I,
    15. Takayama G,
    16. Hizukuri Y,
    17. Ito K,
    18. Taira Y,
    19. Nannya Y,
    20. Tojo A,
    21. Watanabe T,
    22. Tsutsumi S,
    23. Suzuki Y,
    24. Uchimaru K
    : Mechanisms of action and resistance in histone methylation-targeted therapy. Nature 627(8002): 221-228, 2024. DOI: 10.1038/s41586-024-07103-x
    OpenUrlCrossRef
PreviousNext
Back to top

In this issue

In Vivo: 38 (3)
In Vivo
Vol. 38, Issue 3
May-June 2024
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on In Vivo.

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.
Extensive Shrinkage and Long-term Stable Disease in a Teenage Female Patient With High-grade Glioma Treated With Temozolomide and Radiation in Combination With Oral Recombinant Methioninase and a Low-methionine Diet
(Your Name) has sent you a message from In Vivo
(Your Name) thought you would like to see the In Vivo web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
6 + 6 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Extensive Shrinkage and Long-term Stable Disease in a Teenage Female Patient With High-grade Glioma Treated With Temozolomide and Radiation in Combination With Oral Recombinant Methioninase and a Low-methionine Diet
MOTOKAZU SATO, QINGHONG HAN, KOHEI MIZUTA, RYOSUKE MORI, BYUNG MO KANG, SEI MORINAGA, NORITOSHI KOBAYASHI, YASUSHI ICHIKAWA, ATSUSHI NAKAJIMA, ROBERT M. HOFFMAN
In Vivo May 2024, 38 (3) 1459-1464; DOI: 10.21873/invivo.13591

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Extensive Shrinkage and Long-term Stable Disease in a Teenage Female Patient With High-grade Glioma Treated With Temozolomide and Radiation in Combination With Oral Recombinant Methioninase and a Low-methionine Diet
MOTOKAZU SATO, QINGHONG HAN, KOHEI MIZUTA, RYOSUKE MORI, BYUNG MO KANG, SEI MORINAGA, NORITOSHI KOBAYASHI, YASUSHI ICHIKAWA, ATSUSHI NAKAJIMA, ROBERT M. HOFFMAN
In Vivo May 2024, 38 (3) 1459-1464; DOI: 10.21873/invivo.13591
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

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

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • HT1080 Human Fibrosarcoma Cells Selected for Super-eribulin Resistance In Vitro Become More Malignant and Are Arrested Synergistically by Methionine Restriction in Combination With Eribulin in Nude Mice
  • HT1080 Fibrosarcoma With Acquired Trabectedin Resistance: Increased Malignancy But Sustained Sensitivity to Methionine Restriction
  • Ivermectin Combined With Recombinant Methioninase (rMETase) Synergistically Eradicates MiaPaCa-2 Pancreatic Cancer Cells
  • Synergistic Eradication of Fibrosarcoma With Acquired Ifosfamide Resistance Using Methionine Restriction Combined With Ifosfamide in Nude-mouse Models
  • Comparison of Cell-death Kinetics of Recombinant Methioninase (rMETase)-treated Cancer and Normal Cells: Only Cancer Cells Undergo Methionine-depletion Catastrophe at Low rMETase Concentrations
  • Selective Synergy of Recombinant Methioninase Plus Docetaxel Against Docetaxel-resistant and -sensitive Fibrosarcoma Cells Compared to Normal Fibroblasts
  • Recombinant Methioninase Synergistically Reverses High-docetaxel Resistance Developed in Osteosarcoma Cells
  • Recombinant Methioninase Increases Eribulin Efficacy 16-fold in Highly Eribulin-resistant HT1080 Fibrosarcoma Cells, Demonstrating Potential to Overcome the Clinical Challenge of Drug-resistant Soft-tissue Sarcoma
  • First-line Chemotherapy in Combination With Oral Recombinant Methioninase and a Low-methionine Diet for a Stage IV Inoperable Pancreatic-Cancer Patient Resulted in 40% Tumor Reduction and an 86% CA19-9 Biomarker Decrease
  • Overcoming High Trabectedin Resistance of Soft-tissue Sarcoma With Recombinant Methioninase: A Potential Solution of a Recalcitrant Clinical Problem
  • [11C]Methionine PET vs. [18F]Fluorodeoxyglucose PET Whole-body Imaging to Determine the Extent of Methionine-addiction Compared to Glucose-addiction of Primary and Metastatic Cancer of the Trunk in Patients
  • Recombinant Methioninase Is Selectively Synergistic With Doxorubicin Against Wild-type Fibrosarcoma Cells Compared to Normal Cells and Overcomes Highly-Doxorubicin-resistant Fibrosarcoma
  • Induction of the DNA-Repair Gene POLQ only in BRCA1-mutant Breast-Cancer Cells by Methionine Restriction
  • DNA-Binding Agent Trabectedin Combined With Recombinant Methioninase Is Synergistic to Decrease Fibrosarcoma Cell Viability and Induce Nuclear Fragmentation But Not Synergistic on Normal Fibroblasts
  • Google Scholar

More in this TOC Section

  • Surgical Treatment and Prognosis of Soft Tissue Sarcoma in Patients Aged 85 Years and Older
  • Neutrophil-to-Lymphocyte Ratio as a Biomarker for Postoperative Complications in Crohn’s Disease
  • IgA Nephropathy Associated With Infliximab Treatment in Patients With Crohn’s Disease: Study of IgA1 and IgA2 Expression in Glomeruli
Show more Clinical Studies

Similar Articles

Keywords

  • High-grade glioma
  • temozolomide
  • radiation
  • methionine addiction
  • Hoffman effect
  • Methionine restriction
  • oral methioninase
  • low-methionine diet
  • combination therapy
  • synergy
In Vivo

© 2025 In Vivo

Powered by HighWire