Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Dietary methionine influences therapy in mouse cancer models and alters human metabolism

This article has been updated

Abstract

Nutrition exerts considerable effects on health, and dietary interventions are commonly used to treat diseases of metabolic aetiology. Although cancer has a substantial metabolic component1, the principles that define whether nutrition may be used to influence outcomes of cancer are unclear2. Nevertheless, it is established that targeting metabolic pathways with pharmacological agents or radiation can sometimes lead to controlled therapeutic outcomes. By contrast, whether specific dietary interventions can influence the metabolic pathways that are targeted in standard cancer therapies is not known. Here we show that dietary restriction of the essential amino acid methionine—the reduction of which has anti-ageing and anti-obesogenic properties—influences cancer outcome, through controlled and reproducible changes to one-carbon metabolism. This pathway metabolizes methionine and is the target of a variety of cancer interventions that involve chemotherapy and radiation. Methionine restriction produced therapeutic responses in two patient-derived xenograft models of chemotherapy-resistant RAS-driven colorectal cancer, and in a mouse model of autochthonous soft-tissue sarcoma driven by a G12D mutation in KRAS and knockout of p53 (KrasG12D/+;Trp53−/−) that is resistant to radiation. Metabolomics revealed that the therapeutic mechanisms operate via tumour-cell-autonomous effects on flux through one-carbon metabolism that affects redox and nucleotide metabolism—and thus interact with the antimetabolite or radiation intervention. In a controlled and tolerated feeding study in humans, methionine restriction resulted in effects on systemic metabolism that were similar to those obtained in mice. These findings provide evidence that a targeted dietary manipulation can specifically affect tumour-cell metabolism to mediate broad aspects of cancer outcome.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Dietary methionine restriction rapidly and specifically alters methionine and sulfur metabolism and inhibits tumour growth in PDX models of colorectal cancer.
Fig. 2: Dietary methionine restriction sensitizes PDX models of colorectal cancer to chemotherapy with 5-FU.
Fig. 3: Dietary methionine restriction sensitizes mouse models of RAS-driven autochthonous sarcoma to radiation.
Fig. 4: Dietary methionine restriction can be achieved in humans.

Similar content being viewed by others

Data availability

The metabolomics data reported in this study have been deposited in Mendeley Data (https://doi.org/10.17632/zs269d9fvb.1).

Code availability

All computer code is available at: https://github.com/LocasaleLab/Dietary_methionine_restriction.

Change history

  • 23 August 2019

    This article was amended to correct the Peer review information, which was originally incorrect.

References

  1. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    Article  ADS  Google Scholar 

  2. Goncalves, M. D., Hopkins, B. D. & Cantley, L. C. Dietary fat and sugar in promoting cancer development and progression. Annual Rev. Cancer Biol. 3, 255–273 (2019).

    Article  Google Scholar 

  3. Cantor, J. R. et al. Physiologic medium rewires cellular metabolism and reveals uric acid as an endogenous inhibitor of UMP synthase. Cell 169, 258–272 (2017).

    Article  CAS  Google Scholar 

  4. Tardito, S. et al. Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nat. Cell Biol. 17, 1556–1568 (2015).

    Article  CAS  Google Scholar 

  5. Liu, X., Romero, I. L., Litchfield, L. M., Lengyel, E. & Locasale, J. W. Metformin targets central carbon metabolism and reveals mitochondrial requirements in human cancers. Cell Metab. 24, 728–739 (2016).

    Article  CAS  Google Scholar 

  6. Maddocks, O. D. et al. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 493, 542–546 (2013).

    Article  ADS  CAS  Google Scholar 

  7. Maddocks, O. D. K. et al. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 544, 372–376 (2017).

    Article  ADS  CAS  Google Scholar 

  8. Gravel, S. P. et al. Serine deprivation enhances antineoplastic activity of biguanides. Cancer Res. 74, 7521–7533 (2014).

    Article  CAS  Google Scholar 

  9. Kanarek, N. et al. Histidine catabolism is a major determinant of methotrexate sensitivity. Nature 559, 632–636 (2018).

    Article  ADS  CAS  Google Scholar 

  10. Knott, S. R. V. et al. Asparagine bioavailability governs metastasis in a model of breast cancer. Nature 554, 378–381 (2018).

    Article  ADS  CAS  Google Scholar 

  11. Mentch, S. J. et al. Histone methylation dynamics and gene regulation occur through the sensing of one-carbon metabolism. Cell Metab. 22, 861–873 (2015).

    Article  CAS  Google Scholar 

  12. Gao, X., Reid, M. A., Kong, M. & Locasale, J. W. Metabolic interactions with cancer epigenetics. Mol. Aspects Med. 54, 50–57 (2017).

    Article  CAS  Google Scholar 

  13. Orentreich, N., Matias, J. R., DeFelice, A. & Zimmerman, J. A. Low methionine ingestion by rats extends life span. J. Nutr. 123, 269–274 (1993).

    CAS  PubMed  Google Scholar 

  14. Lee, B. C. et al. Methionine restriction extends lifespan of Drosophila melanogaster under conditions of low amino-acid status. Nat. Commun. 5, 3592 (2014).

    Article  ADS  Google Scholar 

  15. Malloy, V. L. et al. Methionine restriction prevents the progression of hepatic steatosis in leptin-deficient obese mice. Metabolism 62, 1651–1661 (2013).

    Article  CAS  Google Scholar 

  16. Ables, G. P., Perrone, C. E., Orentreich, D. & Orentreich, N. Methionine-restricted C57BL/6J mice are resistant to diet-induced obesity and insulin resistance but have low bone density. PLoS ONE 7, e51357 (2012).

    Article  ADS  CAS  Google Scholar 

  17. Malloy, V. L. et al. Methionine restriction decreases visceral fat mass and preserves insulin action in aging male Fischer 344 rats independent of energy restriction. Aging Cell 5, 305–314 (2006).

    Article  CAS  Google Scholar 

  18. Ser, Z. et al. Targeting one carbon metabolism with an antimetabolite disrupts pyrimidine homeostasis and induces nucleotide overflow. Cell Reports 15, 2367–2376 (2016).

    Article  CAS  Google Scholar 

  19. Miousse, I. R. et al. One-carbon metabolism and ionizing radiation: a multifaceted interaction. Biomol. Concepts 8, 83–92 (2017).

    Article  CAS  Google Scholar 

  20. Locasale, J. W. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat. Rev. Cancer 13, 572–583 (2013).

    Article  CAS  Google Scholar 

  21. Hoffman, R. M. & Erbe, R. W. High in vivo rates of methionine biosynthesis in transformed human and malignant rat cells auxotrophic for methionine. Proc. Natl Acad. Sci. USA 73, 1523–1527 (1976).

    Article  ADS  CAS  Google Scholar 

  22. Komninou, D., Leutzinger, Y., Reddy, B. S. & Richie, J. P. Jr. Methionine restriction inhibits colon carcinogenesis. Nutr. Cancer 54, 202–208 (2006).

    Article  CAS  Google Scholar 

  23. Hens, J. R. et al. Methionine-restricted diet inhibits growth of MCF10AT1-derived mammary tumors by increasing cell cycle inhibitors in athymic nude mice. BMC Cancer 16, 349 (2016).

    Article  CAS  Google Scholar 

  24. Guo, H. et al. Therapeutic tumor-specific cell cycle block induced by methionine starvation in vivo. Cancer Res. 53, 5676–5679 (1993).

    CAS  PubMed  Google Scholar 

  25. Saltz, L. B. et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J. Clin. Oncol. 26, 2013–2019 (2008).

    Article  CAS  Google Scholar 

  26. Douillard, J. Y. et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J. Clin. Oncol. 28, 4697–4705 (2010).

    Article  CAS  Google Scholar 

  27. Kirsch, D. G. et al. A spatially and temporally restricted mouse model of soft tissue sarcoma. Nat. Med. 13, 992–997 (2007).

    Article  CAS  Google Scholar 

  28. Moding, E. J. et al. Tumor cells, but not endothelial cells, mediate eradication of primary sarcomas by stereotactic body radiation therapy. Sci. Transl. Med. 7, 278ra34 (2015).

    Article  Google Scholar 

  29. Durando, X. et al. Optimal methionine-free diet duration for nitrourea treatment: a phase I clinical trial. Nutr. Cancer 60, 23–30 (2008).

    Article  CAS  Google Scholar 

  30. Durando, X. et al. Dietary methionine restriction with FOLFOX regimen as first line therapy of metastatic colorectal cancer: a feasibility study. Oncology 78, 205–209 (2010).

    Article  CAS  Google Scholar 

  31. Kim, M. K. et al. Characterization of an oxaliplatin sensitivity predictor in a preclinical murine model of colorectal cancer. Mol. Cancer Ther. 11, 1500–1509 (2012).

    Article  CAS  Google Scholar 

  32. Uronis, J. M. et al. Histological and molecular evaluation of patient-derived colorectal cancer explants. PLoS ONE 7, e38422 (2012).

    Article  ADS  CAS  Google Scholar 

  33. Udofot, O. et al. Pharmacokinetic, biodistribution and therapeutic efficacy of 5-fluorouracil-loaded pH-sensitive PEGylated liposomal nanoparticles in HCT-116 tumor bearing mouse. J. Nat. Sci. 2, e171 (2016).

    PubMed  PubMed Central  Google Scholar 

  34. Lee, C. L. et al. Generation of primary tumors with Flp recombinase in FRT-flanked p53 mice. Dis. Model. Mech. 5, 397–402 (2012).

    Article  CAS  Google Scholar 

  35. Moding, E. J. et al. Atm deletion with dual recombinase technology preferentially radiosensitizes tumor endothelium. J. Clin. Invest. 124, 3325–3338 (2014).

    Article  CAS  Google Scholar 

  36. Liu, X. et al. High-resolution metabolomics with acyl-CoA profiling reveals widespread remodeling in response to diet. Mol. Cell. Proteomics 14, 1489–1500 (2015).

    Article  CAS  Google Scholar 

  37. Liu, X., Ser, Z. & Locasale, J. W. Development and quantitative evaluation of a high-resolution metabolomics technology. Anal. Chem. 86, 2175–2184 (2014).

    Article  CAS  Google Scholar 

  38. Yuan, J., Bennett, B. D. & Rabinowitz, J. D. Kinetic flux profiling for quantitation of cellular metabolic fluxes. Nat. Protocols 3, 1328–1340 (2008).

    Article  CAS  Google Scholar 

  39. Holter, N. S. et al. Fundamental patterns underlying gene expression profiles: simplicity from complexity. Proc. Natl Acad. Sci. USA 97, 8409–8414 (2000).

    Article  ADS  CAS  Google Scholar 

  40. Thiele, I. et al. A community-driven global reconstruction of human metabolism. Nat. Biotechnol. 31, 419–425 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge support from the National Institutes of Health (NIH) R01CA193256, R21CA201963 and P30CA014236 (J.W.L.), R35CA197616 (D.G.K.), T32CA93240 (D.E.C.) and the Canadian Institutes of Health Research (CIHR, 146818) (X.G.). We thank M. L. Kiel and T. Hartman for assistance in designing the diets, and S. Heim for help with food preparation in the human study. The human study was partially supported by the Clinical Research Center at Penn State University (NIH M01RR10732). We gratefully acknowledge members of the Locasale laboratory for discussions and apologize to those whose work we could not cite owing to space constraints.

Author information

Authors and Affiliations

Authors

Contributions

X.G. and J.W.L. designed the study, wrote and edited the paper. D.E.C. and D.G.K. designed the sarcoma experiments and edited the paper. M.L. and D.S.H. designed and implemented the colorectal PDX models and edited the paper. X.G., M.L., D.E.C., G.A. and M.A.R. performed animal experiments. X.G., S.M.S. and M.A.R. performed all cell culture experiments. J.P.R. Jr, A. Ciccarella, A. Calcagnotto and S.N.N. conducted the human study. Z.D. conducted computational analyses with initial help from P.G.M. J.L. and S.J.M. assisted in mass spectrometry metabolomics experiments.

Corresponding author

Correspondence to Jason W. Locasale.

Ethics declarations

Competing interests

J.W.L. and X.G. have patents related to targeting amino acid metabolism in cancer therapy. D.G.K. is a co-founder and has equity in XRAD Therapeutics, a company developing radiosensitizing agents. He also has patents related to radiosensitizing agents.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Peer review information Nature thanks Danica Chen, Naama Kanarek and Alexei Vazquez for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Fig. 1 Dietary restriction of methionine rapidly and specifically alters methionine and sulfur metabolism but maintains overall metabolism in healthy C57BL/6J mice.

a, Dynamic patterns of the top three modes. Standardized concentration (the values are normalized to have mean = 0, s.d. = 1) in mode 1, mode 2 and mode 3. b, Heat map of metabolites in mode 2 and mode 3. PPP, pentose phosphate pathway. c, Volcano plot of metabolites in plasma collected at the end point. P values were determined by two-tailed Student’s t-test. d, Left, pathway analysis of significantly changed (*P < 0.05, two-tailed Student’s t-test) plasma metabolites by 21-day methionine-restricted diet. Right, fold change of altered metabolites in the top three most-affected pathways. Mean ± s.e.m., n = 5 mice per group. *P < 0.05, two-tailed Student’s t-test. e, Relative intensity of plasma amino acids and metabolites in one-carbon metabolism and redox balance at the end of the study. Mean ± s.e.m., n = 5 mice per group. *P < 0.05, two-tailed Student’s t-test. f, Relative intensity of the methionine-metabolism-related metabolites 2-keto-4-methylthiobutyrate and hypotaurine. Mean ± s.d., n = 5 mice per group. *P < 0.05, two-tailed Student’s t-test.

Source data

Extended Data Fig. 2 Dietary restriction of methionine alters methionine metabolism in PDX models of colorectal cancer.

a, Information on original tumours from patients with colorectal cancer. be, Data from the prevention study shown in Fig. 1f. n = 8 mice per group (4 female and 4 male). b, Food intake. Mean ± s.e.m. *P < 0.05, two-tailed Student’s t-test. c, Volcano plots of metabolites in tumour, plasma and liver. P values were determined by two-tailed Student’s t-test. d, Left, Venn diagrams of significantly changed (*P < 0.05, two-tailed Student’s t-test) metabolites in tumour, plasma and liver by methionine restriction and pathway analysis (false discovery rate < 0.5) of the commonly changed metabolites. Right, fold changes of intensity of tumour metabolites in cysteine and methionine metabolism, and taurine and hypotaurine metabolism, induced by methionine restriction. Mean ± s.d. *P < 0.05, two-tailed Student’s t-test. n = 8 mice per group (4 female and 4 male). e, Relative fold change of intensity of amino acids. Mean ± s.d. *P < 0.05, two-tailed Student’s t-test. n = 8 mice per group (4 female and 4 male).

Source data

Extended Data Fig. 3 Methionine restriction leads to specific cell-intrinsic metabolic alterations in tumours.

To determine whether the effect of methionine restriction on tumour growth is systemic, cell autonomous or both, we conducted an integrated analysis of global changes in the metabolic network across tumour, plasma and liver within each model from the prevention study shown in Fig. 1f. n = 8 mice per group (4 female and 4 male). a, Spearman’s rank correlation coefficients of fold change of metabolites in tumour, plasma and liver induced by methionine restriction exhibited strong correlations between each tissue pair, with the highest correlation between the tumour and the plasma in both CRC119 and CRC240. b, Multidimensional scaling analysis of metabolite fold change in response to methionine restriction. In both models, the fold change of metabolites in tumours showed a higher similarity with those in the plasma than with those in the liver. c, d, Liver was the most-affected tissue in both models. c, The effect of methionine restriction on metabolism in tumour, plasma and liver, evaluated by taking the log10 of the fold change. Box limits are the 25th and 75th percentiles, centre line is the median, and the whiskers are the minimal and maximal values. d, Numbers of metabolites significantly altered (*P < 0.05, two-tailed Student’s t-test) by methionine restriction. n = 8 mice per group (4 female and 4 male). e, Schematic defining methionine-related metabolites (metabolized from or to methionine within four reaction steps) and methionine-unrelated metabolites. f, g, A higher proportion of altered metabolites was methionine-related in plasma and tumour compared to liver, in which metabolites altered by methionine restriction were nearly equally distributed between methionine-related and methionine-unrelated groups. f, Fraction of significantly (*P < 0.05, two-tailed Student’s t-test) altered metabolites for methionine-related and methionine-unrelated metabolites in tumour, liver and plasma. g, Numbers of total and significantly altered metabolites for methionine-related and methionine-unrelated metabolites in tumour, liver and plasma. P values were determined by one-sided Fisher’s exact test.

Source data

Extended Data Fig. 4 Methionine restriction inhibits cell proliferation, and most substantially alters cysteine and methionine metabolism in primary colorectal cancer cells.

a, Relative cell numbers in CRC119 and CRC240 primary tumour cells treated with different doses of methionine for 72 h. Mean ± s.d. n = 3 biologically independent samples; similar results were obtained from 3 independent experiments. *P < 0.05, two-tailed Student’s t-test. b, Volcano plots of metabolites in cells cultured in 0 or 100 μM methionine for 24 h. P values were determined by two-tailed Student’s t-test. c, Left, Venn diagram of significantly changed (*P < 0.05, two-tailed Student’s t-test) metabolites in CRC119 and CRC240 primary cells cultured with no methionine versus control (100 μM methionine), and pathway analysis of metabolites that were commonly changed. Right, fold change of metabolites in cysteine and methionine metabolism, and pyrimidine metabolism, in CRC119 and CRC240 primary cells treated with 0 or 100 μM methionine. Mean ± s.d. n = 3 biologically independent samples. *P < 0.05, two-tailed Student’s t-test. d, Relative fold change of intensity of amino acids by methionine deprivation in CRC119 and CRC240 primary cells. Mean ± s.d. n = 3 biologically independent samples. *P < 0.05, two-tailed Student’s t-test.

Source data

Extended Data Fig. 5 Dietary restriction of methionine sensitizes PDX models of colorectal cancer to 5-FU chemotherapy.

a, Volcano plots of metabolites in plasma and liver altered by the combination of dietary restriction of methionine and 5-FU treatment. P values were determined by two-tailed Student’s t-test. b, Effect of 5-FU treatment alone, and a combination of methionine restriction and 5-FU treatment, on metabolites in tumour, plasma and liver, evaluated by taking the log10 of the fold change. Box limits are the 25th and 75th percentiles, centre line is the median, and the whiskers are the minimal and maximal values. The data represents metabolites in liver (337), plasma (282) and tumour (332) from n = 8 mice per group. c, Numbers of metabolites significantly changed by methionine restriction, 5-FU treatment or the combination of methionine restriction and 5-FU treatment in plasma, tumour and liver. *P < 0.05, two-tailed Student’s t-test. d, Pathway analysis of metabolites significantly changed (*P < 0.05, two-tailed Student’s t-test) by methionine restriction, 5-FU treatment or the combination of dietary methionine restriction and 5-FU treatment (false discovery rate < 0.5). eg, Relative intensity of metabolites related to cysteine and methionine metabolism, and nucleotide metabolism, in tumour (e) and redox balance in liver (f) and plasma (g). Mean ± s.e.m. n = 8 mice per group. *P < 0.05, two-tailed Student’s t-test. h, Spearman’s rank correlation coefficients of methionine restriction and 5-FU-induced fold change of metabolites in tumour, plasma and liver from mice on dietary methionine restriction and with 5-FU treatment.

Source data

Extended Data Fig. 6 Inhibition of cell growth mediated by methionine restriction is largely due to interruptions to the production of nucleosides and redox balance.

a, The synergic effects of methionine restriction and 5-FU treatment in CRC119 primary cells and HCT116 cells were evaluated by cell counting. Mean ± s.e.m. n = 3 biological replicates. *P < 0.05 by two-tailed Student’s t-test. b, The rescue effect of choline, formate, homocysteine, homocysteine with vitamin B12, nucleosides and NAC, alone or in combination, on the inhibition of HCT116 cell proliferation mediated by methionine restriction. Mean ± s.e.m., n = 9 biologically independent samples from 3 independent experiments. *P < 0.05 versus control, ^P < 0.05 versus methionine restriction, #P < 0.05 versus 5-FU treatment, $P < 0.05 versus methionine restriction + 5-FU treatment, by two-tailed Student’s t-test. c, Mass intensity for [M + 1] dTTP and [M + 1] methionine in HCT116 cells from the experiment described in Fig. 2h. Mean ± s.d. n = 3 biologically independent samples. *P < 0.05 versus control and #P < 0.05 versus methionine restriction, by two-tailed Student’s t-test.

Source data

Extended Data Fig. 7 Dietary restriction of methionine sensitizes mouse models of RAS-driven autochthonous sarcoma to radiation.

a, Volcano plots of metabolites in tumour, plasma and liver, and pathway analysis of metabolites significantly changed (*P < 0.05, two-tailed Student’s t-test) by dietary restriction of methionine alone (false discovery rate < 1). b, Spearman’s rank correlation coefficients of fold change of metabolites in tumour, plasma and liver induced by methionine restriction. c, Volcano plots of metabolites in tumour, plasma and liver, and pathway analysis of metabolites significantly changed (*P < 0.05, two-tailed Student’s t-test) by dietary restriction of methionine and radiation (false discovery rate < 0.5). d, Spearman’s rank correlation coefficients of fold change of metabolites in tumour, plasma and liver induced by methionine restriction and radiation. e, Relative intensity of metabolites related to cysteine and methionine metabolism, and energy balance in tumours. Mean ± s.d. n = 7 mice per group, except for the methionine-restriction group (n = 6). *P < 0.05 versus control, by two-tailed Student’s t-test. f, g, The largest effects on metabolism occurred in the combination of diet and radiation. f, Effect of methionine restriction and radiation alone, or in combination, on metabolites in tumour, plasma and liver, evaluated by taking the log10 of fold change. Box limits are the 25th and 75th percentiles, centre line is the median, and the whiskers are the minimal and maximal values. The data represent metabolites in liver (319), plasma (308) and tumour (332) from n = 7 mice per group, except for the methionine-restriction group (n = 6). g, Numbers of metabolites significantly changed (*P < 0.05, two-tailed Student’s t-test) by methionine restriction and radiation alone, or in combination.

Source data

Extended Data Fig. 8 Dietary restriction of methionine can be achieved in humans.

a, Heat map of significantly changed (*P < 0.05, two-tailed Student’s t-test) plasma metabolites by dietary intervention, in six human subjects. b, Volcano plot of plasma metabolites. P values were determined by two-tailed Student’s t-test. c, Pathway analysis of altered (*P < 0.05, two-tailed Student’s t-test) plasma metabolites. d, Relative intensity of amino acids in plasma. n = 6 biologically independent humans. *P < 0.05 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 Comparative metabolic effects of methionine restriction across mouse models and humans.

a, Spearman’s rank correlation coefficients of fold changes of methionine-related metabolites induced by methionine restriction (defined in Extended Data Fig. 3f) in plasma samples from non-tumour bearing C57BL/6J mice, CRC119 and CRC240 mouse models, the sarcoma mouse model and healthy human subjects. b, Spearman’s rank correlation coefficients among different models in a, ranked from the highest to the lowest.

Source data

Extended Data Table 1 Methionine concentrations in plasma, tumour and liver across mouse models and humans

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, X., Sanderson, S.M., Dai, Z. et al. Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature 572, 397–401 (2019). https://doi.org/10.1038/s41586-019-1437-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1437-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer