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:

Fasting-mimicking diet and hormone therapy induce breast cancer regression

An Author Correction to this article was published on 04 December 2020

This article has been updated

Abstract

Approximately 75% of all breast cancers express the oestrogen and/or progesterone receptors. Endocrine therapy is usually effective in these hormone-receptor-positive tumours, but primary and acquired resistance limits its long-term benefit1,2. Here we show that in mouse models of hormone-receptor-positive breast cancer, periodic fasting or a fasting-mimicking diet3,4,5 enhances the activity of the endocrine therapeutics tamoxifen and fulvestrant by lowering circulating IGF1, insulin and leptin and by inhibiting AKT–mTOR signalling via upregulation of EGR1 and PTEN. When fulvestrant is combined with palbociclib (a cyclin-dependent kinase 4/6 inhibitor), adding periodic cycles of a fasting-mimicking diet promotes long-lasting tumour regression and reverts acquired resistance to drug treatment. Moreover, both fasting and a fasting-mimicking diet prevent tamoxifen-induced endometrial hyperplasia. In patients with hormone-receptor-positive breast cancer receiving oestrogen therapy, cycles of a fasting-mimicking diet cause metabolic changes analogous to those observed in mice, including reduced levels of insulin, leptin and IGF1, with the last two remaining low for extended periods. In mice, these long-lasting effects are associated with long-term anti-cancer activity. These results support further clinical studies of a fasting-mimicking diet as an adjuvant to oestrogen therapy in hormone-receptor-positive breast cancer.

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: Fasting or FMD potentiates the activity of ET in HR+ BC by reducing circulating growth-promoting factors.
Fig. 2: FMD prevents resistance to combined fulvestrant and palbociclib and reduces tamoxifen-induced endometrial hyperplasia.
Fig. 3: Effects of periodic FMD on disease control and circulating FRFs in patients with HR+ BC and in mice.

Similar content being viewed by others

Data availability

All data generated or analysed during this study are included in this published Article (and its Supplementary Information files). All microarray data are available through the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/) using the accession number GSE121378Source data are provided with this paper.

Change history

References

  1. DeVita, V. J., Laurence, T. S. & Rosenberg, S. A. DeVita, Hellmann and Rosenberg’s Cancer: Principles & Practice of Oncology 11th edn (Wolters Kluwer, 2019).

  2. Araki, K. & Miyoshi, Y. Mechanism of resistance to endocrine therapy in breast cancer: the important role of PI3K/Akt/mTOR in estrogen receptor-positive, HER2-negative breast cancer. Breast Cancer 25, 392–401 (2018).

    Article  PubMed  Google Scholar 

  3. Brandhorst, S. et al. A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab. 22, 86–99 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Di Biase, S. et al. Fasting-mimicking diet reduces HO-1 to promote T cell-mediated tumor cytotoxicity. Cancer Cell 30, 136–146 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Wei, M. et al. Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci. Transl. Med. 9, eaai8700 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. AlFakeeh, A. & Brezden-Masley, C. Overcoming endocrine resistance in hormone receptor-positive breast cancer. Curr. Oncol. 25, S18–S27 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lee, A. V., Cui, X. & Oesterreich, S. Cross-talk among estrogen receptor, epidermal growth factor, and insulin-like growth factor signaling in breast cancer. Clin. Cancer Res. 7, 4429s–4435s (2001).

    CAS  PubMed  Google Scholar 

  8. Sachs, N. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 172, 373–386 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Jones, J. I. & Clemmons, D. R. Insulin-like growth factors and their binding proteins: biological actions. Endocr. Rev. 16, 3–34 (1995).

    CAS  PubMed  Google Scholar 

  10. Garofalo, C., Sisci, D. & Surmacz, E. Leptin interferes with the effects of the antiestrogen ICI 182,780 in MCF-7 breast cancer cells. Clin. Cancer Res. 10, 6466–6475 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Sánchez-Jiménez, F., Pérez-Pérez, A., de la Cruz-Merino, L. & Sánchez-Margalet, V. Obesity and breast cancer: role of leptin. Front. Oncol. 9, 596 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Pollak, M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat. Rev. Cancer 12, 159–169 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Jardé, T., Perrier, S., Vasson, M. P. & Caldefie-Chézet, F. Molecular mechanisms of leptin and adiponectin in breast cancer. Eur. J. Cancer 47, 33–43 (2011).

    Article  PubMed  CAS  Google Scholar 

  15. Saxena, N. K. et al. Concomitant activation of the JAK/STAT, PI3K/AKT, and ERK signaling is involved in leptin-mediated promotion of invasion and migration of hepatocellular carcinoma cells. Cancer Res. 67, 2497–2507 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cristofanilli, M. et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 17, 425–439 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Lasham, A. et al. A novel EGR-1 dependent mechanism for YB-1 modulation of paclitaxel response in a triple negative breast cancer cell line. Int. J. Cancer 139, 1157–1170 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Shajahan-Haq, A. N. et al. EGR1 regulates cellular metabolism and survival in endocrine resistant breast cancer. Oncotarget 8, 96865–96884 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Di Biase, S. et al. Fasting regulates EGR1 and protects from glucose- and dexamethasone-dependent sensitization to chemotherapy. PLoS Biol. 15, e2001951 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Di Leva, G. et al. Estrogen mediated-activation of miR-191/425 cluster modulates tumorigenicity of breast cancer cells depending on estrogen receptor status. PLoS Genet. 9, e1003311 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Hawley, S. A. et al. Phosphorylation by Akt within the ST loop of AMPK-α1 down-regulates its activation in tumour cells. Biochem. J. 459, 275–287 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Arends, J. et al. ESPEN guidelines on nutrition in cancer patients. Clin. Nutr. 36, 11–48 (2017).

    Article  PubMed  Google Scholar 

  23. Grundmann, O., Yoon, S. L. & Williams, J. J. The value of bioelectrical impedance analysis and phase angle in the evaluation of malnutrition and quality of life in cancer patients—a comprehensive review. Eur. J. Clin. Nutr. 69, 1290–1297 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Turner, N. C. et al. Palbociclib in hormone-receptor-positive advanced breast cancer. N. Engl. J. Med. 373, 209–219 (2015).

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  25. Creighton, C. J. et al. Insulin-like growth factor-I activates gene transcription programs strongly associated with poor breast cancer prognosis. J. Clin. Oncol. 26, 4078–4085 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Karey, K. P. & Sirbasku, D. A. Differential responsiveness of human breast cancer cell lines MCF-7 and T47D to growth factors and 17 beta-estradiol. Cancer Res. 48, 4083–4092 (1988).

    CAS  PubMed  Google Scholar 

  27. Baselga, J. et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N. Engl. J. Med. 366, 520–529 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. André, F. et al. Alpelisib for PIK3CA-Mutated, hormone receptor-positive advanced breast cancer. N. Engl. J. Med. 380, 1929–1940 (2019).

    Article  PubMed  Google Scholar 

  29. Hu, R., Hilakivi-Clarke, L. & Clarke, R. Molecular mechanisms of tamoxifen-associated endometrial cancer (Review). Oncol. Lett. 9, 1495–1501 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Piacente, F. et al. Nicotinic acid phosphoribosyltransferase regulates cancer cell metabolism, susceptibility to NAMPT inhibitors, and DNA repair. Cancer Res. 77, 3857–3869 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Caffa, I. et al. Fasting potentiates the anticancer activity of tyrosine kinase inhibitors by strengthening MAPK signaling inhibition. Oncotarget 6, 11820–11832 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Ciribilli, Y. et al. The coordinated p53 and estrogen receptor cis-regulation at an FLT1 promoter SNP is specific to genotoxic stress and estrogenic compound. PLoS One 5, e10236 (2010).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  33. Liu, C. Y. et al. Tamoxifen induces apoptosis through cancerous inhibitor of protein phosphatase 2A-dependent phospho-Akt inactivation in estrogen receptor-negative human breast cancer cells. Breast Cancer Res. 16, 431 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Massarweh, S. et al. Tamoxifen resistance in breast tumors is driven by growth factor receptor signaling with repression of classic estrogen receptor genomic function. Cancer Res. 68, 826–833 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Mishra, A. K., Abrahamsson, A. & Dabrosin, C. Fulvestrant inhibits growth of triple negative breast cancer and synergizes with tamoxifen in ERα positive breast cancer by up-regulation of ERβ. Oncotarget 7, 56876–56888 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Ikeda, H. et al. Combination treatment with fulvestrant and various cytotoxic agents (doxorubicin, paclitaxel, docetaxel, vinorelbine, and 5-fluorouracil) has a synergistic effect in estrogen receptor-positive breast cancer. Cancer Sci. 102, 2038–2042 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Massarweh, S. et al. Mechanisms of tumor regression and resistance to estrogen deprivation and fulvestrant in a model of estrogen receptor-positive, HER-2/neu-positive breast cancer. Cancer Res. 66, 8266–8273 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Vijayaraghavan, S. et al. CDK4/6 and autophagy inhibitors synergistically induce senescence in Rb positive cytoplasmic cyclin E negative cancers. Nat. Commun. 8, 15916 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cook Sangar, M. L. et al. Inhibition of CDK4/6 by palbociclib significantly extends survival in medulloblastoma patient-derived xenograft mouse models. Clin. Cancer Res. 23, 5802–5813 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Michaloglou, C. et al. Combined inhibition of mTOR and CDK4/6 is required for optimal blockade of E2F function and long-term growth inhibition in estrogen receptor-positive breast cancer. Mol. Cancer Ther. 17, 908–920 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lee, C. et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 70, 1564–1572 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ahima, R. S. et al. Role of leptin in the neuroendocrine response to fasting. Nature 382, 250–252 (1996).

    Article  ADS  CAS  PubMed  Google Scholar 

  43. Lee, C. et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci. Transl. Med. 4, 124ra27 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Arends, J. et al. ESPEN expert group recommendations for action against cancer-related malnutrition. Clin. Nutr. 36, 1187–1196 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Reidy, P. T. et al. Protein blend ingestion following resistance exercise promotes human muscle protein synthesis. J. Nutr. 143, 410–416 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rossi, F., Valdora, F., Barabino, E., Calabrese, M. & Tagliafico, A. S. Muscle mass estimation on breast magnetic resonance imaging in breast cancer patients: comparison between psoas muscle area on computer tomography and pectoralis muscle area on MRI. Eur. Radiol. 29, 494–500 (2019).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the Associazione Italiana per la Ricerca sul Cancro (AIRC; IG#17736 and #22098 to A.N.; IG#17605 and IG#21820 to V.D.L.; AIRC Fellowship #22457 to G.S. and V.D.L.; IG#21548 to A.P.; and MFAG#22977 to C.V.), the Fondazione Umberto Veronesi (to A.N., I.C., F.P. and V.D.L.), the Italian Ministry of Health (GR-2011-02347192 to A.N.), the 5 × 1000 2014 Funds to the IRCCS Ospedale Policlinico San Martino (to A.N.), the BC161452 and BC161452P1 grants of the Breast Cancer Research Program (US Department of Defense; to V.D.L. and to A.N., respectively), the US National Institute on Aging–National Institutes of Health (NIA–NIH) grants AG034906 and AG20642 (to V.D.L.), and the Associazione Italiana contro le Leucemie-linfomi e Mieloma (AIL), Sezione Liguria. We thank the High Throughput Screening Facility of the University of Trento (Italy) and T. Bonfiglio (Department of Internal Medicine and Medical Specialties, University of Genoa) for their technical support.

Author information

Authors and Affiliations

Authors

Contributions

A.N. and V.D.L. conceived the study. I.C. and V.S. performed most experiments. P.B., C.Z., E.D., F.P., M.P., G.S. and S.C. performed in vitro experiments. M.W., S.B. and M.C. performed animal work. L.M. and V.G.V. performed the pathology experiments. A.P., S.P., G.Z. and L.F. performed computational and statistical analyses. A.N., C.V., F.V., A.L.C., R.G., C.M., S.G.S., A.A., A.T., A.B. and F.D.B. participated in the clinical trials and collected and analysed clinical data. M.C., P.O., F.M., H.C. and C.V. contributed to the study design. All authors evaluated the results and edited the manuscript. A.N. and V.D.L. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Valter D. Longo or Alessio Nencioni.

Ethics declarations

Competing interests

A.N. and I.C. hold intellectual property rights on clinical uses of fasting-mimicking diets. V.D.L. holds intellectual property rights on clinical uses of fasting-mimicking diets and equity interest in L-Nutra, a company that develops and markets medical food. The remaining authors declare no competing interests.

Additional information

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 Fasting and FMD enhance ET anti-tumour activity in HR+ BC cells.

a, b, MCF7 cells were plated in 96-well plates and treated with STS conditions, tamoxifen or fulvestrant at the indicated concentrations, or their combinations. After 96 h, cells were imaged by light microscopy (b) and their viability was detected (a). c, MCF7 cells were seeded in 6-well plates and cultured with or without STS, tamoxifen (TMX) or fulvestrant (FULV), or their combinations for 24 h. Thereafter, cells were cultured in regular medium for an additional 14 d. Finally, cells were fixed and stained with sulforhodamine B and cell colonies were counted. d, T47D and ZR-75-1 cells were plated in 96-well plates and exposed to STS, TMX at the indicated concentrations or their combination. Cell viability was detected after 96 h. e, ZR-75-1 cells were seeded in 6-well plates and cultured with or without STS, FULV, or their combinations for 24 h before being cultured in regular medium for an additional 14 d. Finally, cells were fixed and stained with sulforhodamine B and cell colonies were counted. f, g, MCF7 xenografts were established in 6–8-week-old female BALB/c nude mice. Once tumours became palpable, mice were randomized to be treated with ad libitum diet (control; n = 5), TMX (n = 5), FULV (n = 5), weekly 48-h water-only fasting (n = 5), or combined TMX + fasting (n = 5) or FULV + fasting (n = 5). Tumour volume and mouse weight were monitored over time (f). At the end of the experiment, mice were killed and tumour masses were imaged and weighed (g). In ae, one representative experiment out of three is presented. In a, c, e,data are from four, three, and four biological replicates (four wells), respectively. In d - left graph, data are from six (left) or four (right) biological replicates (wells).Data are presented as mean ± s.d. (a, ce, g) or s.e.m. (f). Data were analysed by two-tailed Student’s t-test (a, cf; tumour volume at day 32, g) or two-way ANOVA (f).

Source data

Extended Data Fig. 2 Enhancement of ET activity via fasting/FMD in mouse xenografts of HR+ BC cell lines and in human HR+ BC organoids.

a, ZR-75-1 xenograft-bearing, 6–8-week-old female BALB/c nude mice were treated with ad libitum diet (control; n = 5), TMX (n = 5), weekly 48-h FMD (n = 6) or combined TMX and FMD (n = 6). Tumour volume (left) and mouse weight (right) were monitored over time. Mice from all treatment groups were killed at day 65, and tumours were isolated and weighed (middle). b, Growth of T47D xenografts in 6–8-week-old female BALB/c nude mice treated with ad libitum diet (n = 10), TMX (n = 7), fulvestrant (FULV; n = 9), or combined TMX + FMD (n = 5) or FULV + FMD (n = 5). c, Tumour organoids from patients with HR+ BC were cultured with or without TMX, STS conditions or their combination for 120 h, and then were imaged (left; one representative experiment out of three is presented) and their viability quantified (right). Viability of 209M, 213M and 33T BC organoids (graphs at right) was calculated from four biological replicates (wells) in each type of organoid for each treatment condition. d, MCF7 cells were grafted into 6–8-week-old female BALB/c nude mice, and once tumours became palpable, mice were treated with ad libitum diet (n = 14), TMX (n = 14), weekly FMD (n = 18) or their combination (n = 20). Tumour volume (left) and progression-free survival (right) were monitored for each group. n indicates the number of mice (with one tumour per animal; a) or the number of tumours per treatment group (b, d). Data are mean ± s.e.m. (ad) or s.d. (c). Data were analysed by two-way ANOVA (a, left; b; d, left), by two-tailed Student’s t-test (a, right; b, tumour volume at day 105; c) or by log-rank test (d, right).

Source data

Extended Data Fig. 3 FMD-mediated increase in ET anti-cancer activity is mediated by the reduction in circulating insulin, IGF1 and leptin.

a, MCF7 cells were seeded in 96-well plates and cultured for 96 h with or without STS conditions (1% FBS, 0.5 g/l glucose), low-serum conditions (1% FBS, 1 g/l glucose), TMX or FULV at the indicated concentrations, or combinations of these treatments. Thereafter, cell viability was determined. One representative experiment out of three is presented. Cell viability in each treatment condition was calculated from three (MCF7 cells) or four (T47D, ZR-75-1 cells) biological replicates (wells). b, c, Serum β-hydroxybutyrate, IGFBP1, IGFBP3, adiponectin, TNF and IL-1β concentrations in female 6–8-week-old BALB/c nude mice treated with fasting/FMD (or ad libitum diet) with or without TMX or FULV. In all mouse groups, serum was collected at the end of the fasting/FMD. Data are from biological replicates. d, MCF7 cells were injected into 6–8-week-old female BALB/c nude mice; once tumours became palpable, mice were randomized to be treated with ad libitum diet (n = 6), FULV (n = 6), FULV + weekly FMD (n = 6), FULV + FMD + i.p. insulin (n = 5), FULV + FMD + IGF1 (n = 5), FULV + FMD + leptin (n = 5), or FULV + FMD + combined insulin, IGF1 and leptin (FRFs; n = 7). FRF administration was withdrawn at day 35 (crossover), while it was started in mice that had only been treated with FULV + FMD. Right, tumour volume in each treatment group at day 57; n, number of tumours per treatment group. e, MCF7 cells were seeded in 96-well plates and cultured for 96 h with or without STS, 10 μM FULV, insulin, IGF1, leptin (at the indicated concentrations), combined insulin, IGF1 and leptin, or combinations of these treatments. Cells were then imaged by light microscopy (left) and cell viability was determined (right). One representative experiment out of three is presented. f, At the end of the experiment shown in d, tumour masses were isolated for protein lysate generation. Total and phosphorylated AKT (Ser473) and p70S6K (Thr389) and vinculin (on the same gel) were assessed by immunoblotting. One representative experiment out of three (n = 5 or 6 tumour masses/treatment group were evaluated) is presented. For gel source data, see Supplementary Fig. 1. Data are from biological replicates and represent mean ± s.d. (ac, e, right) or s.e.m. (d). Data were analysed by two-tailed Student’s t-test.

Source data

Extended Data Fig. 4 Fasting or FMD and oestrogen therapy cooperate to inhibit PI3K–AKT–mTOR and oestrogen receptor signalling in HR+ BC cells.

a, b, MCF7, T47D and ZR-75-1 cells were seeded in 6-well plates and cultured for 48 h with or without STS conditions in the presence or absence of tamoxifen (5 μM) or fulvestrant (10 μM). Thereafter, cells were subjected to protein lysate generation, and PI3K–AKT–mTOR signalling and EGR1, PTEN and β-actin (on the same gel) levels were detected by immunoblotting. For gel source data, see Supplementary Fig. 1. In b, protein bands were quantified and normalized to vinculin levels; data are from biological replicates and were obtained by three different experiments. c, MCF7 cells were injected into 6–8-week-old female BALB/c nude mice. Once tumours became palpable, mice were randomized to be treated with ad libitum diet (n = 3), FULV (n = 3), TMX (n = 3), weekly 48-h FMD (n = 4), FULV + FMD (n = 4) or TMX + FMD (n = 4). Mice were killed at the end of the fourth FMD cycle. Tumour masses were isolated and EGR1 and PTEN expression were detected by qPCR. Data are from biological replicates. n, number of tumours isolated and used for this experiment. d, Metastasis-derived HR+ BC organoids were cultured with or without TMX, STS or their combination for 48 h. Thereafter, organoids were isolated and subjected to RNA isolation, and EGR1 and PTEN expression was quantified by qPCR. Data are from biological replicates. One representative experiment out of three is presented. e, MCF7, T47D and ZR-75-1 cells were seeded in 96-well plates and cultured for 48 h with or without 1 μg/ml cycloheximide (CYCLO), STS conditions, TMX (5 μM), FULV (10 μM) or their combinations. Thereafter, protein synthesis was detected by OPP assay. Left, cells were imaged by fluorescence microscopy (one representative image out of three biological replicates (wells) is shown). Right, single-cell analysis was performed by acquiring the fluorescence signal from >1,000 cells per treatment condition (range 11548290). In be, data represent mean ± s.d. P values were calculated by two-tailed Student’s t-test. b, ns: non-significant; *P < 0.05; **P < 0.01; ***P < 0.001 (for P values, see Source Data file).

Source data

Extended Data Fig. 5 EGR1, PTEN and reduced AKT activation mediate the cooperation between ET and fasting/FMD in BC cells.

ad, MCF7 cells were transduced with either one of two independent EGR1-targeting shRNAs (#1 or #2). In a, d, cells were seeded in 96-well plates and cultured with or without STS conditions, TMX or FULV at the indicated concentrations (5 μM TMX or 10 μM FULV in a) or their combinations for 96 h and then cell viability was detected (a, d). In a (upper panel), b, c, cells were plated in 6-well plates and cultured with or without STS, 5 μM TMX, 10 μM FULV or their combinations for 48 h. Afterwards, cells were subjected to protein lysate generation, and EGR1, PTEN and phosphorylated (Ser473) and total AKT, as well as GAPDH, were detected by immunoblotting. e, MCF7 cells transduced with a control vector (n = 12), an EGR1- (n = 10) or a PTEN-targeting (n = 10) shRNA, or myr-AKT (n = 11) were injected into 6–8-week-old female BALB/c nude mice. Once tumours became palpable, mice were treated with FULV and weekly cycles of FMD, and tumour volume was monitored. n, number of tumours per treatment group. f, MCF7 cells engineered to express either a control scrambled shRNA or an EGR1-targeting shRNA (shRNA#1) were seeded in 96-well plates. Twenty-four hours later, cells were cultured with or without TMX (5 μM), STS, GDC0068 (1 μM), AZD5363 (500 nM), LY294002 (2 μM) or their combinations. Cell viability was detected after 96 h. g, h, MCF7 cells were transduced with either a PTEN-targeting shRNA or myr-AKT. Cells were plated in 6-well plates and subjected to protein lysate generation. PTEN (g), AKT (h) and vinculin (g, h) were detected by immunoblotting. Thereafter, cells were seeded in 96-well plates and cultured with or without STS, TMX (5 μM), FULV (10 μM) or their combinations for 96 h and then cell viability was detected. i, MCF7 cells were seeded in 6-well plates and 24 h later were cultured with or without STS + TMX (5 μM), STS + TMX + combined insulin (400 pM), IGF1 (5 ng/ml) and leptin (50 mg/ml) or STS + TMX + 17β-oestradiol (100 nM). Forty-eight hours later, cells were subjected to protein lysate generation and EGR1, PTEN, 4E-BP1 and vinculin were detected by immunoblotting. j, k, MCF7 cells that were engineered with either a control vector or myr-AKT were cultured for 48 h with or without TMX (5 μM), FULV (10 μM), STS or their combinations. Thereafter, cells were subjected to protein lysate generation and monitoring of EGR1 and GAPDH levels by immunoblotting (j) or for RNA extraction and EGR1 and PTEN mRNA quantification (k). In a (lower panel), d, f, g (lower panel), h (lower panel), k, data are from biological replicates (in d, data are from four biological replicates (wells) per treatment condition). In a (upper inset), b, c, g (upper inset), h (upper inset), i, j, one representative experiment out of three is presented. Loading controls (GAPDH, vinculin) were always run on the same gels that were used to detect other proteins. For gel source data, see Supplementary Fig. 1. Data are presented as mean ± s.d. (a, d, fh, k) or s.e.m. (e). In a, d, fh, k, P values were determined by two-tailed Student’s t-test. In e, data were analysed by two-way ANOVA with Bonferroni post-hoc test and by two-tailed Student’s t-test (tumour volumes at day 40).

Source data

Extended Data Fig. 6 STS conditions cooperate with ET to activate AMPK and to dampen oestrogen receptor transcriptional activity in BC cells.

a, MCF7 cells transduced with a control vector, a PTEN-targeting shRNA or myr-AKT were plated into 6-well plates and cultured with or without STS conditions, TMX (5 μM), FULV (10 μM) or their combinations for 48 h. Afterwards, cells were subjected to protein lysate generation, and total and phosphorylated AMPK (Thr172) were detected by immunoblotting. The loading control for the immunoblots with PTEN-silenced MCF7 cells (vinculin) is shown in Supplementary Fig. 1. The loading control for the immunoblots with myr-AKT-expressing MCF7 (GAPDH) is shown in Extended Data Fig. 5j. Both loading controls were run on the same gels that were used to detect total and phosphorylated AMPK. For gel source data, see Supplementary Fig. 1. b, MCF7 cells were plated into 24-well plates and transfected with a pGL3 promoter plasmid or with a pS2/TFF1 reporter vector containing 1.3 kb of the proximal promoter of the oestrogen-responsive gene TFF1 cloned in the pGL3-basic backbone. Afterwards, cells were cultured with or without STS, TMX (5 μM), FULV (10 μM) or their combinations for 48 h. Finally, luciferase reporter activity was measured. c, MCF7, T47D and ZR-75-1 cells were plated in 6-well plates and cultured with or without STS, TMX (5 μM) or their combinations for 48 h. Thereafter, cells were subjected to RNA extraction, and PGR, TFF1 and GREB1 expression was detected by qPCR. In ac, one representative experiment out of three is presented. In b, c, data are from biological replicates and are presented as mean ± s.d. P values were determined by two-tailed Student’s t-test.

Source data

Extended Data Fig. 7 FMD cooperates with ETs to induce cell cycle arrest in HR+ BC.

ac, MCF7 cells were plated in 6-well plates and cultured with or without STS conditions, 5 μM tamoxifen (TMX) or their combination. After 24 h, RNA was isolated and used for gene expression microarray experiments (three biological replicates for each treatment condition were used). GSEA, represented by a heat map (a), was done by performing 10,000 permutations and using the REACTOME Pathways data set. Box plots (b) were generated using the ggplot2 package; to evaluate the differences between the groups, a non-parametric two-sided Wilcoxon test was used. The box-plot centre lines indicate the median value. c, Enrichment scores for G1-specific transcription genes of STS + TMX versus CTR, STS, or TMX groups. NES: normalized enrichment score. d, MCF7, T47D and ZR-75-1 cells were treated with TMX (5 μM), STS or their combinations. After 48 h, cells were subjected to RNA extraction or protein lysate generation. E2F1, E2F2, CCND1 and CCNE1 mRNA expression was detected by qPCR. e, CCND1, phosphorylated (Ser807/811) and total RB protein and vinculin (on the same gel) were detected by immunoblotting in the MCF7 cells. f, Tumour masses from mice that were treated for 4 weeks with ad libitum diet (n = 5), FULV (n = 4), weekly 48 h FMD (n = 4) or their combination (n = 4) were isolated at the end of the last FMD cycle. CCND1 and vinculin (run on the same gel) were detected by immunoblotting (left). CCND1 levels were quantified and normalized to vinculin (right). n signifies the number of tumours isolated per treatment group. g, T47D and ZR-75-1 cells were cultured in vitro for 48 h with or without TMX (5 μM), STS conditions or their combination, and then used to generate protein lysates. Phosphorylated (Ser807/811) RB protein bands were quantified and normalized to total RB. In d, f (right), g, data are from biological replicates. In g (right), data were obtained through three independent experiments. In df (left), one representative experiment out of three (d, e) or out of two (f, left) is presented. In d, f, (right), g, data are presented as mean ± s.d. P values were determined by two-tailed Student’s t-test. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 8 Combined FMD and ET downregulate CCND1 via EGR1 upregulation and AKT inhibition and revert acquired resistance to fulvestrant plus palbociclib.

a, b, MCF7, T47D and ZR-75-1 cells were plated into 96-well plates and cultured with or without TMX (5 μM) or FULV (10 μM), STS conditions or their combinations for 48 h. They were then subjected to cell cycle analysis by propidium iodide staining of isolated cell nuclei and flow cytometry. c, Upper panel, MCF7 cells were transduced with either a control scrambled shRNA or an EGR1-targeting shRNA (EGR1-shRNA#1); lower panel, MCF7 cells were transduced with a control vector or myr-AKT. Cells were cultured with or without TMX (5 μM) or FULV (10 μM), STS or their combinations for 48 h. Afterwards, cells were subjected to protein lysate generation, and CCND1 and vinculin (on the same gel) levels were detected by immunoblotting. For gel source data, see Supplementary Fig. 1. d, MCF7 cells transduced with a control vector or myr-AKT were cultured with or without TMX (5 μM) or FULV (10 μM), STS or their combinations for 48 h and then subjected to cell cycle analysis. e, MCF7 cells were injected orthotopically into 6–8-week-old female NOD/SCIDγ mice. Once tumours became palpable, mice were treated with ad libitum diet (n = 15), FULV (n = 16), cyclic FMD (n = 15), palbociclib (n = 15), FULV + PALB (n = 18), FULV + FMD (n = 16), PALB + FMD (n = 10) or FULV + PALB + FMD (n = 18), and their progression-free survival was monitored over time. f, g, MCF7 cells with acquired resistance to combined FULV + PALB (from Fig. 2b) were isolated and expanded ex vivo. Resistant MCF7 as well as parental MCF7 cells (control) were injected into 6–8-week-old female NOD/SCIDγ mice. Once tumours became palpable, mice were randomly assigned to be treated with or without FULV + PALB (mice with control MCF7 treated with vehicle: n = 5, with FULV + PALB: n = 4; mice with resistant MCF7 treated with vehicle: n = 4, with FULV + PALB: n = 5). Tumour volumes were monitored, and at the end of the experiment (day 38) tumour masses were isolated and subjected to histology (haematoxylin and eosin, HE) and immunohistochemical detection of Ki67+ cells (g). In g, right, 3–5 slices were prepared from each tumour (technical replicates) and subjected to Ki67+ cell enumeration. In e, f, n indicates the number of mice per treatment group (one tumour per mouse). In ad, one representative experiment out of three is presented. Data are mean ± s.e.m. (f) or mean ± s.d. (g, right). In e, progression-free survival was analysed by log-rank test. In f, data were analysed by two-way ANOVA with Bonferroni post-hoc test and by two-tailed Student’s t-test (day 38). In g, P values were determined by two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 Fasting or FMD prevents tamoxifen-induced endometrial hyperplasia and reduces intra-abdominal fat.

ad, Six–eight-week-old female BALB/c mice were treated for 5 weeks with ad libitum diet (= 11), tamoxifen (TMX; n = 11), weekly 48-h water-only fasting (n = 11) or FMD (n = 8), TMX + fasting (n = 11) or TMX + FMD (n = 8). Mice from all treatment groups were killed at the end of the last fasting/FMD cycle. Uteri were collected, imaged (a), fixed for histology (b) and subjected to protein lysate generation and RNA extraction. In addition, intra-abdominal fat depots were also isolated. Tff1, Pten and Egr1 mRNA levels in mice uteri were determined by qPCR (c), and total and phosphorylated AKT (Ser473), EGR1, PTEN and vinculin in mouse uteri were detected by immunoblotting (d; vinculin was detected on the same gel as EGR1 and PTEN). For gel source data, see Supplementary Fig. 1. e, Intra-abdominal fat depots isolated from the mice were imaged. In c, d, data are from biological replicates. P values were determined by two-tailed Student’s t-test.

Source data

Extended Data Fig. 10 Effects of periodic FMD on circulating FRFs and on disease in patients with HR+ BC and in HR+ BC mouse xenografts.

a, Body weight, hand grip, phase angle, fat-free and fat mass in patients (n = 23) with HR+ BC treated with ET and cyclic FMD in the NCT03595540 clinical trial. To evaluate changes in these parameters, we fitted a linear mixed-effects model taking into account absolute values as a function of time with a random covariate represented by subject ID. b, Quantification of total muscle area (highlighted in red) at L3 level (patient 1) and at the Louis angle level (patient 3) in patients with HR+ BC treated with ET and cyclic FMD (NCT03595540). c, Serum IGFBP1 and IFGBP3 (n = 6), blood glucose (n = 7) and ketone bodies (n = 11) before and after an FMD cycle in patients with BC treated with ET and FMD in the NCT03595540 trial. d, Serum leptin, adiponectin, C-peptide, IGF1, IGFBP1 and IFGBP3 (n = 7) and blood glucose (n = 12) before and after an FMD cycle in patients with  BC treated with ET and FMD in the NCT03340935 clinical trial. e, Serum C-peptide, IGFBP1 and IGFBP3 in patients with HR+ BC treated with ET and cyclic FMD before and 3 weeks after an FMD cycle (n = 23; NCT03595540 trial). f, 6–8-week-old BALB/c nude mice were treated with ad libitum diet (n = 7), fulvestrant (n = 7), tamoxifen (n = 6), weekly FMD (n = 7), FULV + FMD (n = 6) or TMX + FMD (n = 7) for 2 weeks; 1 week after the end of the last FMD cycle, mice were killed, serum was isolated and serum C-peptide, IGF1 and leptin were measured by ELISA. g, h, MCF7 cells were injected into 6–8-week-old female BALB/c nude mice. Once tumours became palpable, mice were randomly assigned to be treated for 1 month with ad libitum diet (g, n = 9; h, n = 8), TMX (g, n = 9; h, n = 7), FULV (g, h, n = 9), FMD (g, n = 7), TMX + FMD (g, n = 9), FULV + FMD (g, n = 9), 48 h water-only fasting (h, n = 8), TMX + fasting (h, n = 14) or FULV + fasting (h, n = 10), followed by observation. Tumour volume (h) and mouse survival (g, h) were monitored. n, number of mice per treatment group (f, g) or of tumours per treatment group (h). In cf, data are from biological replicates. In f, data are mean ± s.d. In h (left), data are mean ± s.e.m. In ce, data were analysed by two-tailed Student’s paired t-test. In f, P values were determined by two-tailed Student’s t-test. In g, h (right), data were analysed by log-rank test. In h (left), results were analysed by two-way ANOVA with Bonferroni post-hoc test.

Source data

Supplementary information

Supplementary Figure 1

This file contains the uncropped blots with size marker indications.

Reporting Summary

Supplementary Tables

This file contains Supplementary Tables 1-3. The Supplementary Tables include: clinical characteristics, adverse events and best response of the patients with HR+ BC who were treated with ET plus periodic FMD within the clinical trials NCT03595540 and NCT03340935; the complete blood counts and a metabolic panel post-FMD in patients with HR+ BC treated with endocrine therapy and periodic FMD in the clinical trial NCT03595540; the list of primers that were utilized for the qPCR reactions of this study.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caffa, I., Spagnolo, V., Vernieri, C. et al. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 583, 620–624 (2020). https://doi.org/10.1038/s41586-020-2502-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2502-7

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