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.

  • Article
  • Published:

Low glycaemic diets alter lipid metabolism to influence tumour growth

Abstract

Dietary interventions can change metabolite levels in the tumour microenvironment, which might then affect cancer cell metabolism to alter tumour growth1,2,3,4,5. Although caloric restriction (CR) and a ketogenic diet (KD) are often thought to limit tumour progression by lowering blood glucose and insulin levels6,7,8, we found that only CR inhibits the growth of select tumour allografts in mice, suggesting that other mechanisms contribute to tumour growth inhibition. A change in nutrient availability observed with CR, but not with KD, is lower lipid levels in the plasma and tumours. Upregulation of stearoyl-CoA desaturase (SCD), which synthesises monounsaturated fatty acids, is required for cancer cells to proliferate in a lipid-depleted environment, and CR also impairs tumour SCD activity to cause an imbalance between unsaturated and saturated fatty acids to slow tumour growth. Enforcing cancer cell SCD expression or raising circulating lipid levels through a higher-fat CR diet confers resistance to the effects of CR. By contrast, although KD also impairs tumour SCD activity, KD-driven increases in lipid availability maintain the unsaturated to saturated fatty acid ratios in tumours, and changing the KD fat composition to increase tumour saturated fatty acid levels cooperates with decreased tumour SCD activity to slow tumour growth. These data suggest that diet-induced mismatches between tumour fatty acid desaturation activity and the availability of specific fatty acid species determine whether low glycaemic diets impair tumour growth.

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

Access options

Buy this article

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

Fig. 1: CR, but not the KD, impairs growth of PDAC allograft tumours.
Fig. 2: CR and the KD differentially alter nutrient levels in the plasma and TIF.
Fig. 3: Increased SCD activity is required for cancer cells to adapt to exogenous lipid limitation.
Fig. 4: Low glycaemic diets impair tumour SCD, which interacts with changes in lipid availability to affect tumour growth.

Similar content being viewed by others

Data availability

All data generated or analysed in this study are included in the published article and in Supplementary Table 1, Supplementary Figs. 123 and Source Data for Figs. 14 and Extended Data Figs. 18. Correspondence and requests for materials should be addressed to Matthew G. Vander Heiden (mvh@mit.edu). Source data are provided with this paper.

References

  1. Lien, E. C. & Vander Heiden, M. G. A framework for examining how diet impacts tumour metabolism. Nat. Rev. Cancer 19, 651–661 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Sullivan, M. R. et al. Increased serine synthesis provides an advantage for tumors arising in tissues where serine levels are limiting. Cell Metab. 29, 1410–1421 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sullivan, M. R. et al. Quantification of microenvironmental metabolites in murine cancers reveals determinants of tumor nutrient availability. Elife 8, e44235 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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  PubMed  Google Scholar 

  5. Gao, X. et al. Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature 572, 397–401 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kalaany, N. Y. & Sabatini, D. M. Tumours with PI3K activation are resistant to dietary restriction. Nature 458, 725–731 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hopkins, B. D. et al. Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 560, 499–503 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Nencioni, A., Caffa, I., Cortellino, S. & Longo, V. D. Fasting and cancer: molecular mechanisms and clinical application. Nat. Rev. Cancer 18, 707–719 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Danai, L. V. et al. Altered exocrine function can drive adipose wasting in early pancreatic cancer. Nature 558, 600–604 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gocheva, V. et al. Quantitative proteomics identify Tenascin-C as a promoter of lung cancer progression and contributor to a signature prognostic of patient survival. Proc. Natl Acad. Sci. USA 114, E5625–E5634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kennedy, A. R. et al. A high-fat, ketogenic diet induces a unique metabolic state in mice. Am. J. Physiol. Endocrinol. Metab. 292, E1724–E1739 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Mukherjee, P., El-Abbadi, M. M., Kasperzyk, J. L., Ranes, M. K. & Seyfried, T. N. Dietary restriction reduces angiogenesis and growth in an orthotopic mouse brain tumour model. Br. J. Cancer 86, 1615–1621 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhang, J. et al. Low ketolytic enzyme levels in tumors predict ketogenic diet responses in cancer cell lines in vitro and in vivo. J. Lipid Res. 59, 625–634 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hosios, A. M., Li, Z., Lien, E. C. & Vander Heiden, M. G. Preparation of lipid-stripped serum for the study of lipid metabolism in cell culture. Bio Protoc. 8, e2876 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Peck, B. & Schulze, A. Lipid desaturation—the next step in targeting lipogenesis in cancer. FEBS J. 283, 2767–2778 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Vriens, K. et al. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature 566, 403–406 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  17. Kim, W. et al. Polyunsaturated fatty acid desaturation is a mechanism for glycolytic NAD+ recycling. Cell Metab. 29, 856–870 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Titov, D. V. et al. Complementation of mitochondrial electron transport chain by manipulation of the NAD+/NADH ratio. Science 352, 231–235 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Diehl, F. F., Lewis, C. A., Fiske, B. P. & Vander Heiden, M. G. Cellular redox state constrains serine synthesis and nucleotide production to impact cell proliferation. Nat. Metab. 1, 861–867 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Piccolis, M. et al. Probing the global cellular responses to lipotoxicity caused by saturated fatty acids. Mol. Cell 74, 32–44 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mauvoisin, D. & Mounier, C. Hormonal and nutritional regulation of SCD1 gene expression. Biochimie 93, 78–86 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Young, R. M. et al. Dysregulated mTORC1 renders cells critically dependent on desaturated lipids for survival under tumor-like stress. Genes Dev. 27, 1115–1131 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ackerman, D. et al. Triglycerides promote lipid homeostasis during hypoxic stress by balancing fatty acid saturation. Cell Rep. 24, 2596–2605 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kamphorst, J. J. et al. Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc. Natl Acad. Sci. USA 110, 8882–8887 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Coulston, A. M. The role of dietary fats in plant-based diets. Am. J. Clin. Nutr. 70, 512S–515S (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Pascual, G. et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 541, 41–45 (2017).

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Siri-Tarino, P. W., Chiu, S., Bergeron, N. & Krauss, R. M. Saturated fats versus polyunsaturated fats versus carbohydrates for cardiovascular disease prevention and treatment. Annu. Rev. Nutr. 35, 517–543 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mayers, J. R. et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 353, 1161–1165 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lewis, C. A. et al. Tracing compartmentalized NADPH metabolism in the cytosol and mitochondria of mammalian cells. Mol. Cell 55, 253–263 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Heinrich, P. et al. Correcting for natural isotope abundance and tracer impurity in MS-, MS/MS- and high-resolution-multiple-tracer-data from stable isotope labeling experiments with IsoCorrectoR. Sci. Rep. 8, 17910 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Antoniewicz, M. R., Kelleher, J. K. & Stephanopoulos, G. Measuring deuterium enrichment of glucose hydrogen atoms by gas chromatography/mass spectrometry. Anal. Chem. 83, 3211–3216 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Argus, J. P. et al. Development and application of FASA, a model for quantifying fatty acid metabolism using stable isotope labeling. Cell Rep. 25, 2919–2934 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vichai, V. & Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 1, 1112–1116 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Colditz, G. A. & Hankinson, S. E. The Nurses’ Health Study: lifestyle and health among women. Nat. Rev. Cancer 5, 388–396 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Belanger, C. F., Hennekens, C. H., Rosner, B. & Speizer, F. E. The nurses’ health study. Am. J. Nurs. 78, 1039–1040 (1978).

    CAS  PubMed  Google Scholar 

  38. Health Professionals Follow-Up Study (Harvard T. H. Chan School of Public Health, 2020); https://sites.sph.harvard.edu/hpfs/

  39. Hu, F. B. et al. Reproducibility and validity of dietary patterns assessed with a food-frequency questionnaire. Am. J. Clin. Nutr. 69, 243–249 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Feskanich, D. et al. Reproducibility and validity of food intake measurements from a semiquantitative food frequency questionnaire. J. Am. Diet Assoc. 93, 790–796 (1993).

    Article  CAS  PubMed  Google Scholar 

  41. Willett, W. C. et al. Reproducibility and validity of a semiquantitative food frequency questionnaire. Am. J. Epidemiol. 122, 51–65 (1985).

    Article  CAS  PubMed  Google Scholar 

  42. Rimm, E. B. et al. Reproducibility and validity of an expanded self-administered semiquantitative food frequency questionnaire among male health professionals. Am. J. Epidemiol. 135, 1114–1126 (1992).

    Article  CAS  PubMed  Google Scholar 

  43. Willett, W. C. Nutritional Epidemiology (Oxford University Press, 2012).

  44. Yuan, C. et al. Validity of a dietary questionnaire assessed by comparison with multiple weighed dietary records or 24-hour recalls. Am. J. Epidemiol. 185, 570–584 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Yuan, C. et al. Relative validity of nutrient intakes assessed by questionnaire, 24-hour recalls, and diet records as compared with urinary recovery and plasma concentration biomarkers: findings for women. Am. J. Epidemiol. 187, 1051–1063 (2018).

    Article  PubMed  Google Scholar 

  46. Halton, T. L., Liu, S., Manson, J. E. & Hu, F. B. Low-carbohydrate-diet score and risk of type 2 diabetes in women. Am. J. Clin. Nutr. 87, 339–346 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Halton, T. L. et al. Low-carbohydrate-diet score and the risk of coronary heart disease in women. N. Engl. J. Med. 355, 1991–2002 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Liu, Y. et al. Plant-based and animal-based low-carbohydrate diets and risk of hepatocellular carcinoma among US men and women. Hepatology 73, 175–185 (2020).

    Article  Google Scholar 

  49. Stampfer, M. J. et al. Test of the National Death Index. Am. J. Epidemiol. 119, 837–839 (1984).

    Article  CAS  PubMed  Google Scholar 

  50. Rich-Edwards, J. W., Corsano, K. A. & Stampfer, M. J. Test of the National Death Index and Equifax Nationwide Death Search. Am. J. Epidemiol. 140, 1016–1019 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Colditz, G. A. et al. Validation of questionnaire information on risk factors and disease outcomes in a prospective cohort study of women. Am. J. Epidemiol. 123, 894–900 (1986).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Vander Heiden laboratory for useful discussions and experimental advice. E.C.L. was supported by the Damon Runyon Cancer Research Foundation (DRG-2299-17). A.N.L. was a Robert Black Fellow of the Damon Runyon Cancer Research Foundation (DRG-2241-15) and was supported by an NIH Pathway to Independence Award (K99CA234221). Z.L. and K.M.S. were supported by NIH training grant T32GM007287. B.M.W. was supported by the Lustgarten Foundation, the Dana-Farber Cancer Institute Hale Family Center for Pancreatic Cancer Research, the NIH (U01CA210171 and P50CA127003), Stand Up To Cancer, the Pancreatic Cancer Action Network, the Noble Effort Fund, the Wexler Family Fund, Promises for Purple and the Bob Parsons Fund. M.G.V.H. acknowledges support from the Emerald Foundation, the Lustgarten Foundation, a Faculty Scholar grant from the Howard Hughes Medical Institute, Stand Up To Cancer, the MIT Center for Precision Cancer Medicine, the Ludwig Center at MIT and the NIH (R35CA242379, R01CA168653, R01CA201276 and P30CA14051). The Nurses’ Health Study and the Health Professionals Follow-up Study were supported by grants UM1 CA186107, U01 CA176726, P01 CA87969, U01 CA167552 and R03 CA223619 from the NIH. The authors thank all participants and staff of the Nurses’ Health Study and the Health Professionals Follow-Up Study for their contributions to this research. We are grateful for help from the following state cancer registries: Alabama, Arizona, Arkansas, California, Colorado, Connecticut, Delaware, Florida, Georgia, Idaho, Illinois, Indiana, Iowa, Kentucky, Louisiana, Maine, Maryland, Massachusetts, Michigan, Nebraska, New Hampshire, New Jersey, New York, North Carolina, North Dakota, Ohio, Oklahoma, Oregon, Pennsylvania, Rhode Island, South Carolina, Tennessee, Texas, Virginia, Washington and Wyoming. The authors also thank the Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School for leading the Nurses’ Health Study and the Harvard T.H. Chan School of Public Health for leading the Health Professionals Follow-Up Study. The authors assume full responsibility for analyses and interpretation of these data.

Author information

Authors and Affiliations

Authors

Contributions

E.C.L. and M.G.V.H. conceived the project. E.C.L. performed the experiments and analysed data. A.M.W. and A.N.L. assisted with animal experiments and interpreted data. A.M.W., Z.L. and K.M.S. assisted with fatty acid profiling experiments and interpreted data. Y.Z., C.Y. and B.M.W. performed analyses of patient population data from the Nurses’ Health Study and the Health Professionals Follow-up Study. E.C.L. and M.G.V.H. wrote the manuscript, with input from all authors.

Corresponding author

Correspondence to Matthew G. Vander Heiden.

Ethics declarations

Competing interests

M.G.V.H. is a scientific advisor for Agios Pharmaceuticals, Aeglea Biotherapeutics, iTeos Therapeutics and Auron Therapeutics. B.M.W. declares research funding from Celgene and Eli Lilly and consulting for BioLineRx, Celgene and GRAIL. E.C.L., A.M.W., Y.Z., C.Y., Z.L., A.N.L. and K.M.S. declare no competing interests.

Additional information

Peer review information Nature thanks Valter Longo, Costas Lyssiotis, Stephan Herzig and the other, anonymous, reviewer(s) for their contribution to the peer review of this work .

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

Extended data figures and tables

Extended Data Fig. 1 Caloric restriction, but not the ketogenic diet, impairs growth of tumor allografts.

ad, Normalized tumor volumes (a, c) and normalized tumor weights (b, d) of subcutaneous AL1376 PDAC allografts in male mice exposed to CR or a control diet (a, b), or the KD or a control diet (c, d). Tumor volumes and weights normalized to animal body weight are shown. (a, b) Control n = 5 mice, CR n = 4 mice; (c, d) Control n = 5 mice, KD n = 5 mice. eh, Tumor volumes (e, g) and tumor weights (f, h) of subcutaneous AL1376 PDAC allografts in female mice exposed to CR or a control diet (e, f), or the KD or a control diet (g, h). (e, f) Control n = 8 mice, CR n = 8 mice; (g, h) Control n = 4 mice, KD n = 4 mice. il, Tumor volumes (i, k) and tumor weights (j, l) of subcutaneous LGSP non-small cell lung cancer (NSCLC) allografts in male (i, j) and female (k, l) mice exposed to CR or a control diet. (i, j) Control n = 10 mice, CR n = 11 mice; (k, l) Control n = 5 mice, CR n = 5 mice. mp, Tumor volumes (m, o) and tumor weights (n, p) of subcutaneous LGSP NSCLC allografts in male (m, n) and female (o, p) mice exposed to KD or a control diet. (m, n) Control n = 5 mice, KD n = 5 mice; (o, p) Control n = 3 mice, KD n = 3 mice. Data are presented as box-and-whisker plots displaying median, interquartile range (boxes), and minima and maxima (whiskers) (b, d, f, h, j, l, n, p) or mean ± SEM (a, c, e, g, i, k, m, o). Comparisons were made using a two-tailed Student’s t-test (b, d, f, h, j, l, n, p) or two-way repeated measures analysis of variance (ANOVA) (a, c, e, g, i, k, m, o)

Source data.

Extended Data Fig. 2 Caloric restriction and the ketogenic diet have distinct effects on whole-body physiology.

a, b, Daily food consumption by weight (a) and daily caloric consumption (b) of the control diet and CR by male and female mice. c, Body weights of male and female mice exposed to CR or a control diet. Male Control n = 10, Male CR n = 11, Female Control n = 5, Female CR n = 5. d, e, Ki67 (d) and cleaved caspase 3 (CC3) (e) immunohistochemical staining in subcutaneous AL1376 tumors from mice exposed to a control or CR diet. n = 3 tumors per group. 5-8 independent fields from each tumor were quantified: Control n = 23, CR n = 18 (d); n = 17 per group (e). f, Weights of gastrocnemius muscle (Gastroc) and white adipose tissue (WAT) in male and female mice exposed to CR or a control diet. Tissue weights normalized to animal body weight are shown. n = 5 per group. gi, Plasma glucagon (g), FGF21 (h), and corticosterone (i) levels in male mice exposed to CR or a control diet. Control n = 6, CR n = 5. j, k, Daily food consumption by weight (j) and daily caloric consumption (k) of the control diet and KD by male and female mice. Male Control n = 5, Male KD n = 5, Female Control n = 4, Female KD n = 5. l, Body weights of male and female mice exposed to KD or a control diet. Male Control n = 5, Male KD n = 5, Female Control n = 3, Female KD n = 3. m, n, Ki67 (m) and CC3 (n) immunohistochemical staining in subcutaneous AL1376 tumors from mice exposed to a control diet or the KD. n = 3 tumors per group. 5-8 independent fields from each tumor were quantified: n = 24 per group (m, n). o, Weights of Gastroc and WAT in male and female mice exposed to KD or a control diet. Tissue weights normalized to animal body weight are shown. Male Control n = 5, Male KD n = 5, Female Control n = 6, Female KD n = 6. pr, Plasma glucagon (p), FGF21 (q), and corticosterone (r) levels in male mice exposed to KD or a control diet. (p) n = 4 per group; (qr) n = 5 per group. Data are presented as mean ± SEM (ac, fl, or) or box-and-whisker plots displaying median, interquartile range (boxes), and minima and maxima (whiskers) (d, e, m, n). Comparisons were made using a two-tailed Student’s t-test

Source data.

Extended Data Fig. 3 β-OHB is metabolized by PDAC cells.

a, Concentrations of β-OHB in plasma and tumor interstitial fluid (TIF) from mice implanted with subcutaneous AL1376 tumors exposed to a control diet, CR, or the KD. Plasma Control (CR cohort) n = 8, Plasma CR n = 8, Plasma Control (KD cohort) n = 10, Plasma KD n = 9, TIF Control (CR cohort) n = 16, TIF CR n = 13, TIF Control (KD cohort) n = 5, TIF KD n = 6. b, Doubling times of AL1376 cells cultured in media with or without 5 mM β-OHB. c, Fractional labeling of [M+2] TCA cycle metabolites from AL1376 cells cultured in the presence of 5 mM [U-13C]-β-OHB for 24 h. d, Mass isotopologue distribution of labeled palmitate from AL1376 cells cultured in the presence of 5 mM [U-13C]-β-OHB for 24 h. e, f, The indicated fatty acid levels (e) and MUFA/SFA ratios (f) measured in AL1376 cells cultured in media with or without 5 mM β-OHB for 24 h. Data are presented as box-and-whisker plots displaying median, interquartile range (boxes), and minima and maxima (whiskers) (a) or mean ± SEM (bf). Unless otherwise indicated, n = 3 biologically independent replicates, and comparisons were made using a two-tailed Student’s t-test

Source data.

Extended Data Fig. 4 Increased stearoyl-CoA desaturase activity is required for cancer cells to adapt to exogenous lipid limitation.

a, Levels of the indicated fatty acids in lipidated and de-lipidated fetal bovine serum (FBS). b, Doubling times of the specified cells cultured in media containing lipidated versus de-lipidated serum. c, Fold changes in levels of the indicated fatty acids in cells cultured in de-lipidated versus lipidated media for 24 h. d, Relative fatty acid synthesis rates in the indicated cells cultured in lipidated versus de-lipidated media. A one-tailed Student’s t-test was used for comparison between groups. e, f, Representative mass isotopologue distributions (MIDs) of 16:0 (e) and 18:0 (f) from AL1376 cells labeled with [U-13C]-glucose and [U-13C]-glutamine for 24 h in lipidated versus de-lipidated media. g, h, Representative MIDs of 16:1(n-7) (g) and 18:1(n-9) (h) from AL1376 cells labeled with [U-13C]-glucose and [U-13C]-glutamine for 24 h in lipidated versus de-lipidated media containing 50 nM A939572. MIDs above M+12 are predominantly observed, consistent with a majority of lipogenic acetyl-CoA being derived from glucose and glutamine. i, Immunoblot for the mature activated form of SREBP1 (m-SREBP1) and β-actin in lysates from AL1376 cells cultured in lipidated versus de-lipidated media for 24 h, with or without 50 nM A939572, 50 µM 18:1(n-9), or 50 µM 18:2(n-6). Data is representative of three biologically independent experiments. j, Absolute fatty acid concentrations in AL1376 and LGSP cells cultured in lipidated media. k, l, Ratios of 16:1(n-7)/16:0 (k) and 18:1(n-9)/18:0 (l) in cells cultured in lipidated versus de-lipidated media with or without the SCD inhibitor A939572 (LGSP: 50 nM; HeLa, Panc1, A549: 200 nM) for 24 h. m, n, 16:1(n-7) (m) and 18:1(n-9) (n) synthesis rates in cells cultured in lipidated versus de-lipidated media with or without A939572 (LGSP: 50 nM; HeLa, Panc1, A549: 200 nM) for 24 h as indicated. A one-tailed ratio paired t-test was used for comparison between groups. o, Doubling times of the specified cells cultured in lipidated versus de-lipidated media with the indicated concentrations of A939572. p, Doubling times of LGSP cells cultured in de-lipidated media with or without 50 nM A939572, BSA, 100 µM 18:1(n-9), 100 µM 18:2(n-6), or 15 µM 16:0. q, Doubling times of cells cultured in de-lipidated media containing 200 nM A939572, BSA, or 25 µM 18:2(n-6). rv, 18:1(n-9) (r), 16:1(n-7) (s), 18:1(n-9)/18:0 (t), 16:1(n-7)/16:0 (u), and 16:0 (v) levels in LGSP cells cultured in de-lipidated media with or without 50 nM A939572, BSA, 100 µM 18:1(n-9), 100 µM 18:2(n-6), or 15 µM 16:0 for 48 h. w, Relative fatty acid synthesis rates in LGSP cells cultured in de-lipidated media containing 50 nM A939572, BSA, 100 µM 18:1(n-9), or 100 µM 18:2(n-6). x, Doubling times of cells cultured in de-lipidated media containing A939572 (LGSP: 50 nM; HeLa, Panc1, A549: 200 nM), the FASN inhibitor GSK2194069 (LGSP: 0.3 µM; HeLa, A549: 0.015 µM; Panc1: 0.05 µM), or the ACC inhibitor PF-05175157 (LGSP: 10 µM; HeLa: 0.3 µM; Panc1: 1 µM; A549: 0.5 µM). A paired two-tailed Student’s t-test was used for comparison between groups in Panc1 and A549 cells. All data are presented as mean ± SEM; unless otherwise indicated, n = 3 biologically independent replicates, and a two-tailed Student’s t-test was used for comparison between groups unless otherwise noted above

Source data.

Extended Data Fig. 5 Determining whether SCD inhibition affects cancer cell proliferation via NAD+ limitation or changes to cellular fatty acid levels.

a, Doubling times of the indicated cell lines cultured in de-lipidated media with or without 1 mM pyruvate and rotenone (AL1376: 400 nM; LGSP, A549: 100 nM; HeLa, Panc1: 50 nM). b, Doubling times of the indicated cell lines cultured in de-lipidated media containing the indicated concentrations of the SCD inhibitor A939572 and 1 mM pyruvate. c, Doubling times of the indicated cell lines cultured in de-lipidated media with or without 50 nM A939572. di, Relative levels of 16:0 (d), 18:0 (e), 16:1(n-7) (f), 18:1(n-9) (g), 16:1(n-7)/16:0 (h), and 18:1(n-9)/18:0 (i) in the indicated cell lines cultured in de-lipidated media with or without 50 nM A939572. jl, Relative levels of 16:1(n-10) (j), normalized levels of 16:1(n-10)/16:0 (k), and relative levels of 16:1(n-10)/16:0 (l) in the indicated cell lines cultured in de-lipidated media with or without 50 nM A939572. All data are presented as mean ± SEM; n = 3 biologically independent replicates. A two-tailed Student’s t-test was used for comparison between groups

Source data.

Extended Data Fig. 6 Caloric restriction and the ketogenic diet alter fatty acid composition and SCD activity in tumors.

a, The indicated fatty acid levels measured in subcutaneous AL1376 tumors from mice exposed to a control or CR diet. Control n = 10, CR n = 8. bd, The indicated fatty acid levels (b, d) and MUFA/SFA ratios (c) measured in subcutaneous AL1376 tumors from male or female mice exposed to a control or CR diet. Male Control n = 6, Male CR n = 4, Female Control n = 4, Female CR n = 4. eg, The indicated fatty acid levels (e, g) and MUFA/SFA ratios (f) measured in subcutaneous LGSP tumors from male mice exposed to a control or CR diet. Control n = 6, CR n = 6. h-i, Immunoblot and quantification for mouse Scd1 (mSCD1) and vinculin in subcutaneous AL1376 tumors from female mice (h, n = 7 per group) or subcutaneous LGSP tumors from male mice (i, n = 6 per group) exposed to a control or CR diet. j, The indicated fatty acid levels measured in subcutaneous AL1376 tumors from mice exposed to a control diet or KD. Control n = 8, KD n = 9. km, The indicated fatty acid levels (k, m) and MUFA/SFA ratios (l) measured in subcutaneous AL1376 tumors from male or female mice exposed to a control diet or KD. Male Control n = 5, Male KD n = 5, Female Control n = 3, Female KD n = 4. np, The indicated fatty acid levels (n, p) and MUFA/SFA ratios (o) measured in subcutaneous LGSP tumors from male mice exposed to a control diet or KD. Control n = 6, KD n = 6. qr, Immunoblot and quantification for mSCD1 and vinculin in subcutaneous AL1376 tumors from female mice (q, Control n = 3, KD n = 4) or subcutaneous LGSP tumors from male mice (r, n = 6 per group) exposed to a control diet or KD. s, AL1376 and LGSP cells were serum-starved in the presence of 1% Lipid Mixture for 16 h followed by insulin stimulation (100 nM) for 3 h. Lysates were immunoblotted for mSCD1, p-AKT S473, AKT, and vinculin. Data is representative of three biologically independent experiments. Data are presented as box-and-whisker plots displaying median, interquartile range (boxes), and minima and maxima (whiskers) (a, eg, j, np) or mean ± SEM (bd, h, i, km, qr). Comparisons were made using a two-tailed Student’s t-test

Source data.

Extended Data Fig. 7 Exogenous SCD expression partially rescues tumor growth inhibition by caloric restriction.

a, b, Tumor volumes (a) and tumor weights (b) normalized to animal body weight of subcutaneous AL1376 tumors expressing EV or SCD-HA in male mice exposed to a control or CR diet. EV: Control n = 4 mice, EV: CR n = 4 mice, SCD-HA: Control n = 4 mice, SCD-HA: CR n = 4 mice. c, d, Tumor volumes (c) and tumor weights (d) of subcutaneous AL1376 tumors expressing EV or SCD-HA in female mice exposed to a control or CR diet. EV: Control n = 4 mice, EV: CR n = 4 mice, SCD-HA: Control n = 4 mice, SCD-HA: CR n = 4 mice. e, f, Tumor volumes (e) and tumor weights (f) normalized to animal body weight of subcutaneous AL1376 tumors expressing EV or SCD-HA in female mice exposed to a control or CR diet. EV: Control n = 4 mice, EV: CR n = 4 mice, SCD-HA: Control n = 4 mice, SCD-HA: CR n = 4 mice. g, h, Daily food consumption by weight (g) and daily caloric consumption (h) of the control diet and CR by male and female mice. i, j, Tumor weights (i) and tumor weights normalized to animal body weight (j) of subcutaneous LGSP tumors expressing EV or SCD-HA in mice exposed to a control or CR diet. EV: Control n = 6 mice, EV: CR n = 5 mice, SCD-HA: Control n = 6 mice, SCD-HA: CR n = 6 mice. k, Immunoblot for HA, human SCD (hSCD), mouse Scd1 (mSCD1), and vinculin in subcutaneous AL1376 tumors expressing an empty vector (EV) or SCD-HA from mice exposed to a control or CR diet for 18 days. ln, The indicated MUFA/SFA ratios (l) and fatty acid levels (m, n) measured in subcutaneous AL1376 tumors expressing EV or SCD-HA from mice exposed to a control or CR diet for 18 days. EV: Control n = 6, EV: CR n = 6, SCD-HA: Control n = 6, SCD-HA: CR n = 6. o, Immunoblot for HA, hSCD, mSCD1, and vinculin in subcutaneous AL1376 tumors expressing an empty vector (EV) or SCD-HA from mice exposed to a control or CR diet for 7 days. pr, The indicated MUFA/SFA ratios (p) and fatty acid levels (qr) measured in subcutaneous AL1376 tumors expressing EV or SCD-HA from mice exposed to a control or CR diet for 7 days. EV: Control n = 6, EV: CR n = 6, SCD-HA: Control n = 6, SCD-HA: CR n = 6. Data are presented as box-and-whisker plots displaying median, interquartile range (boxes), and minima and maxima (whiskers) (b, d, f, i, j, ln, pr) or mean ± SEM (a, c, e, g, h). Comparisons were made using two-way repeated measures analysis of variance (ANOVA) (a, c, e) or a two-tailed Student’s t-test (b, d, f, i, j, ln, pr)

Source data.

Extended Data Fig. 8 Changing the fat composition of caloric restriction and the ketogenic diet alters tumor growth and tumor fatty acid composition.

a, b, The indicated fatty acid levels measured in plasma from mice exposed to a control diet, CR, high fat caloric restriction diet consisting of soybean oil (HFCR-Soybean), and high fat caloric restriction diet consisting of palm oil (HFCR-Palm). Control n = 6, CR n = 5, HFCR-Soybean n = 5, HFCR-Palm n = 5. ce, The indicated fatty acid levels (c, e) and MUFA/SFA ratios (d) measured in subcutaneous AL1376 tumors from mice exposed to a control diet, CR, HFCR-Soybean, or HFCR-Palm. Control n = 7, CR n = 7, HFCR-Soybean n = 7, HFCR-Palm n = 7. f, g, Tumor volumes (f) and tumor weights (g) of subcutaneous LGSP tumors in mice exposed to a control diet, CR, HFCR-Soybean, or HFCR-Palm. Control n = 5 mice, CR n = 6 mice, HFCR-Soybean n = 5 mice, HFCR-Palm n = 6 mice. hj, The indicated fatty acid levels (h, j) and MUFA/SFA ratios (i) measured in subcutaneous LGSP tumors from mice exposed to a control diet, CR, HFCR-Soybean, or HFCR-Palm. Control n = 6, CR n = 6, HFCR-Soybean n = 6, HFCR-Palm n = 6. k, Tumor weights of subcutaneous LGSP tumors expressing an empty vector (EV) or SCD-HA in mice exposed to a control or KD diet consisting of palm oil (KD-Palm). EV: Control n = 6 mice, EV: KD-Palm n = 6 mice, SCD-HA: Control n = 6 mice, SCD-HA: KD-Palm n = 6 mice. ln, The indicated fatty acid levels measured in plasma from mice exposed to a control diet or KD-Palm. Control n = 12, KD-Palm n = 12. or, The indicated fatty acid levels (o, p, r) and MUFA/SFA ratios (q) measured in subcutaneous AL1376 tumors expressing an EV or SCD-HA from mice exposed to a control diet or KD-Palm. EV: Control n = 6, EV: KD-Palm n = 7, SCD-HA: Control n = 6, SCD-HA: KD-Palm n = 6. Data are presented as box-and-whisker plots displaying median, interquartile range (boxes), and minima and maxima (whiskers) (ae, gr) or mean ± SEM (f). Comparisons were made using a two-tailed Student’s t-test (ae, hr), a two-way repeated measures analysis of variance (ANOVA) (f), or a one-way ANOVA (g)

Source data.

Extended Data Table 1 Compositions of diets used in this study
Extended Data Table 2 Low-carbohydrate dietary pattern or macronutrient intake and overall survival among 1,165 pancreatic cancer patients in two large cohorts

Supplementary information

Supplementary Figs.

Supplementary Figs. 1–23, containing uncropped scans of western blots.

Reporting Summary

Peer Review File

Supplementary Table 1

Raw data used for fatty acid source analysis (FASA) synthesis rate calculations in Fig. 3 and Extended Data Fig. 4 .

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lien, E.C., Westermark, A.M., Zhang, Y. et al. Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature 599, 302–307 (2021). https://doi.org/10.1038/s41586-021-04049-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04049-2

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