Liver Metastatic Breast Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , ,

The median overall survival of patients with metastatic breast cancer is only 2–3 years, and for patients with untreated liver metastasis, it is as short as 4–8 months. Improving the survival of women with breast cancer requires more effective anti-cancer strategies, especially for metastatic dis- ease. Nutrients can influence tumor microenvironments, and cancer metabolism can be manipulated via dietary modification to enhance anti-cancer strategies. Yet, there are no standard evidence-based recommendations for diet therapies before or during cancer treatment, and few studies provide definitive data that certain diets can mediate tumor progression or therapeutic effectiveness in human cancer. This review focuses on metastatic breast cancer, in particular liver metastatic forms, and recent studies on the impact of diets on disease progression and treatment.

  • breast cancer liver metastasis
  • western diet
  • fasting-mimicking diet

1. Introduction

Data from the American Cancer Society estimate that there will be 1.9 million new cancer cases diagnosed and 608,570 cancer deaths in the US in 2021 [1]. For women in the US, breast cancer is the most common cancer (30% of all new cases), with an estimated 281,550 newly diagnosed cases and 43,600 deaths in 2021 [1]. In 2018, an estimated 3.7 million women were living with breast cancer in the US [2]. Furthermore, global breast cancer mortality is increasing substantially, especially in developing regions such as Latin America and the Caribbean, rising by an estimated 7 million deaths every five years [3]. These trends demonstrate a need for continued efforts to abate a serious public health concern.

One emerging approach to intervening in breast cancer outcomes is the use of targeted dietary interventions. Indeed, accumulating data indicates that practical clinical dietary interventions, such as the ketogenic diet, can improve the efficacy of anticancer therapy [4]. Thus, dietary approaches hold the potential to enhance therapeutic effectiveness and improve overall survival in breast cancer patients, thereby offering new promise for clinical practice that can change outcomes for a substantial number of patients worldwide.

Here, we review studies demonstrating how diet impacts disease progression and treatment in metastatic breast cancer, particularly metastases to the liver.

2. Breast Cancer Metastasis

Approximately 63% of breast cancer patients are diagnosed with local-stage breast cancer, 27% with the regional-stage disease, and 6% with distant (metastatic) disease [1]. In the US, an estimated >168,000 women were living with metastatic breast cancer in 2020 [5]. Al- though metastatic disease accounts for a small percentage of breast cancer cases, metastatic tumors are responsible for more than 90% of all cancer-related deaths [6]. Indeed, among breast cancer cases, the five-year survival rate for those with localized disease is more than 90%, but for those with metastases, the rate falls to just 28% [7]. Furthermore, the median survival of patients with metastatic disease at the time of diagnosis is approximately 18–24 months, and roughly 13% will survive 10 years [8]. About one-third of women diagnosed early with non-metastatic breast cancer will ultimately develop metastatic disease [9], which tends to develop resistance to therapies [10]. These phenomena underscore the increasing importance of developing therapies to prevent and treat metastatic disease and thus improve the overall survival of women with breast cancer [6].

The sites of distant metastasis among stage IV breast cancer patients include bone (68.8%), lung (16.0%), liver (13.3%), and brain (1.9%) [11]. Based on limited therapy options and dire disease outcomes for patients with liver metastasis, we will focus on liver metastatic ER+ breast cancer in this review. Important data on the impact of the location of metastases on patient survival come from the National Cancer Institute’s Surveillance, Epidemiology, and End Results (SEER), a network of tumor registries that include about 30% of the US population and harboring data from 1975 to 2017 [9]. Of the 2.4 million cancer patients within this database, 5.14% present with synchronous liver metastases (LM) [9,12]. Half of all breast cancer patients develop LM, which often carries poor survival [13]—as low as 4–8 months if the disease is left untreated [14]. Surprisingly, metastatic breast cancer in the liver is observed more frequently in younger women (occurring in 34.2% of all patients < 50 years) than in older women (occurring in 8.9% of all patients ≥ 50 years) [15,16]. In addition, patients with hormone receptor (HR)+/HER2+ breast cancer with LM have longer median survival than patients with HR+/HER2- and triple-negative breast cancer due to the introduction of HER2-targeted therapy [17,18]. Thus, liver metastatic disease represents an important subgroup of breast cancer diagnoses that warrants focused efforts to improve outcomes.

3. Breast Cancer Liver Metastasis Diagnosis, Therapies, and Potential Treatments

Breast cancer LM may at first be asymptomatic, but possible symptoms include fatigue and weakness, pain or discomfort in the mid-section, weight loss or poor appetite, swelling in the legs, fever, and/or a yellow tint to the skin or whites of the eyes [19]. It is often identified by liver function tests that detect liver disease or damage [20]. Diagnosis may also be facilitated through imaging (MRI (magnetic resonance imaging), CT (computed tomography), PET (positron emission tomography), and PET/CT) or biopsy [21].

Most patients with breast cancer LM are treated with either systemic medications or local treatment [22]. Chemotherapy, hormonal therapies, or targeted therapies are common systemic treatments [23]. Chemotherapy involves the use of anti-cancer drugs to destroy or damage cancer cells [24]. Hormonal therapies use drugs such as tamoxifen, aromatase inhibitors, and fulvestrant to target estrogen and help shrink or slow the growth of HR+ metastatic breast cancer [25–27]. Targeted therapies exploit specific characteristics of cancer cells to treat metastatic disease. Some common targeted therapeutics are everolimus, bevacizumab+paclitaxel, palbociclib, and ribociclib [28–33]. Table 1 describes more current options such as potential oral selective estrogen receptor degraders or other pathway inhibitors. Local treatments for breast cancer LM include surgery, radiation therapy, and local chemotherapy. Surgery is most often used when the liver is the only site of metastasis and the symptoms are severe. Radiation therapies such as stereotactic body radiation therapy and Y-90 (Yttrium 90) radioembolization deliver or target radiation therapy directly to tumors in the liver [34,35].

Endocrine therapies reduce breast cancer mortality and relieve symptoms, but some persistent tumor cells frequently develop resistance in the metastatic and adjuvant setting [36–38]. Liver metastatic estrogen receptor α (ERα)-positive breast cancer is currently incurable [39]. Some potential small molecule therapies show good tumor responses in metastatic breast cancers. Axl kinase is associated with aggressive migratory behavior in tumors in a mouse model, and a combination of R428, a selective small molecule Axl inhibitor, with cisplatin positively reinforces both agents to block liver micro-metastases [40]. VERU-111 acts by depolymerizing microtubules, often leading to cell apoptosis due to the inability to complete mitosis, and is highly effective, especially against fibrous tumors and metastases [41]. Recent evidence indicates that ErSO, a small molecule activator of a stress response mechanism that stimulates the anticipatory unfolded protein response (a-UPR), can eradicate most lung, bone, and liver metastases in orthotopic cell line xenograft and patient-derived xenograft (PDX) mouse models [39].

Table 1. Selected Oral Selective Estrogen Receptor Degraders or Other Inhibitors in Clinical Investigation.

This entry is adapted from the peer-reviewed paper 10.3390/nu14122376

References

  1. 1. Tarver, T. Breast Cancer Facts & Figures 2021; American Cancer Society: Atlanta, GA, USA, 2021.2. National Cancer Institute. Cancer Stat Facts: Female Breast Cancer; National Cancer Institute: Bethesda, MD, USA, 2020.3. Azamjah, N.; Soltan-Zadeh, Y.; Zayeri, F. Global Trend of Breast Cancer Mortality Rate: A 25-Year Study. Asian Pac. J. Cancer Prev. 2019, 20, 2015–2020. [CrossRef] [PubMed]4. Lévesque, S.; Pol, J.G.; Ferrere, G.; Galluzzi, L.; Zitvogel, L.; Kroemer, G. Trial watch: Dietary interventions for cancer therapy. OncoImmunology 2019, 8, e1591878. [CrossRef]5. Mariotto, A.B.; Etzioni, R.; Hurlbert, M.; Penberthy, L.; Mayer, M. Estimation of the Number of Women Living with Metastatic Breast Cancer in the United States. Cancer Epidemiol. Prev. Biomark. 2017, 26, 809–815. [CrossRef] [PubMed]6. Gupta, G.P.; Massagué, J. Cancer metastasis: Building a framework. Cell 2006, 127, 679–695.[CrossRef] [PubMed]7. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [CrossRef] [PubMed]8. Eng, L.G.; Dawood, S.; Sopik, V.; Haaland, B.; Tan, P.S.; Bhoo-Pathy, N.; Warner, E.; Iqbal, J.; Narod, S.A.; Dent, R. Ten-year survival in women with primary stage IV breast cancer. Breast Cancer Res. Treat. 2016, 160, 145–152. [CrossRef] [PubMed]9. Howlader, N.; Noone, A.M.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.R.; et al. SEER Cancer Statistics Review, 1975–2017; National Cancer Institute: Bethesda, MD, USA, 2020. Available online: https://seer.cancer.gov/csr/1975_2017/ (accessed on 5 April 2022).10. Soni, D.A.; Ren, Z.; Hameed, O.; Chanda, D.; Morgan, C.J.; Siegal, G.P.; Wei, S. Breast Cancer Subtypes Predispose the Site of Distant Metastases. Am. J. Clin. Pathol. 2015, 143, 471–478. [CrossRef]11. Gong, Y.; Liu, Y.-R.; Ji, P.; Hu, X.; Shao, Z.-M. Impact of molecular subtypes on metastatic breast cancer patients: A SEER population-based study. Sci. Rep. 2017, 7, 45411. [CrossRef]12. Horn, S.R.; Stoltzfus, K.C.; Lehrer, E.J.; Dawson, L.A.; Tchelebi, L.; Gusani, N.J.; Sharma, N.K.; Chen, H.; Trifiletti, D.M.; Zaorsky,N.G. Epidemiology of liver metastases. Cancer Epidemiol. 2020, 67, 101760. [CrossRef] [PubMed]13. Rashid, N.S.; Grible, J.M.; Clevenger, C.V.; Harrell, J.C. Breast cancer liver metastasis: Current and future treatment approaches. Clin. Exp. Metastasis 2021, 38, 263–277. [CrossRef] [PubMed]14. Adam, R.; Aloia, T.; Krissat, J.; Bralet, M.P.; Paule, B.; Giacchetti, S.; Delvart, V.; Azoulay, D.; Bishmuth, H.; Castaing, D. Is liver resection justified for patients with hepatic metastases from breast cancer? Ann. Surg. 2006, 244, 897. [CrossRef] [PubMed]15. De Ridder, J.; De Wilt, J.H.W.; Simmer, F.; Overbeek, L.; Lemmens, V.; Nagtegaal, I. Incidence and origin of histologically confirmed liver metastases: An explorative case-study of 23,154 patients. Oncotarget 2016, 7, 55368–55376. [CrossRef] [PubMed]16. Cummings, M.C.; Simpson, P.T.; Reid, L.E.; Jayanthan, J.; Skerman, J.; Song, S.; Reed, A.E.M.; Kutasovic, J.R.; Morey, A.L.; Marquart, L.; et al. Metastatic progression of breast cancer: Insights from 50 years of autopsies. J. Pathol. 2014, 232, 23–31.[CrossRef] [PubMed]17. Ji, L.; Cheng, L.; Zhu, X.; Gao, Y.; Fan, L.; Wang, Z. Risk and prognostic factors of breast cancer with liver metastases. BMC Cancer 2021, 21, 1–15. [CrossRef]18. Xie, J.Z.; Xu, A. Population-Based Study on Liver Metastases in Women with Newly Diagnosed Breast Cancer. Cancer Epidemiol Biomark. Prev. 2019, 28, 283–292. [CrossRef]19. Diamond, J.R.; Finlayson, C.A.; Borges, V.F. Hepatic complications of breast cancer. Lancet Oncol. 2009, 10, 615–621. [CrossRef]20. Patanaphan, V.; Salazar, O.M.; Risco, R. Breast cancer: Metastatic patterns and their prognosis. South. Med. J. 1988, 81, 1109–1112. [CrossRef]21. Cao, R.; Wang, L.-P. Serological Diagnosis of Liver Metastasis in Patients with Breast Cancer. Cancer Biol. Med. 2012, 9, 57–62. [CrossRef]22. Bale, R.; Putzer, D.; Schullian, P. Local Treatment of Breast Cancer Liver Metastasis. Cancers 2019, 11, 1341. [CrossRef]23. Higgins, M.J.; Baselga, J. Targeted therapies for breast cancer. J. Clin. Investig. 2011, 121, 3797–3803. [CrossRef]24. Carrick, S.; Parker, S.; Thornton, C.E.; Ghersi, D.; Simes, J.; Wilcken, N. Single agent versus combination chemotherapy for metastatic breast cancer. Cochrane Database Syst. Rev. 2009, 2021, CD003372. [CrossRef]25. Osborne, C.K. Tamoxifen in the treatment of breast cancer. N. Engl. J. Med. 1998, 339, 1609–1618. [CrossRef] [PubMed]26. Smith, I.E.; Dowsett, M. Aromatase inhibitors in breast cancer. N. Engl. J. Med. 2003, 348, 2431–2442. [CrossRef] [PubMed]27. Spoerke, J.M.; Gendreau, S.; Walter, K.; Qiu, J.; Wilson, T.R.; Savage, H.; Aimi, J.; Derynck, M.K.; Chen, M.; Chan, I.T.; et al. Heterogeneity and clinical significance of ESR1 mutations in ER-positive metastatic breast cancer patients receiving fulvestrant. Nat. Commun. 2016, 7, 1–10. [CrossRef]28. Mayer, I.A.; Abramson, V.G.; Formisano, L.; Balko, J.M.; Estrada, M.V.; Sanders, M.E. A phase Ib study of alpelisib (BYL719), a PI3Kα-specific inhibitor, with letrozole in ER+/HER2− metastatic breast cancer. Clin. Cancer Res. 2017, 23, 26–34. [CrossRef]29. Coleman, R.L.; Brady, M.F.; Herzog, T.J.; Sabbatini, P.; Armstrong, D.K.; Walker, J.L.; Kim, B.G.; Fujiwara, K.; Tewari, K.S.; O’Malley, D.M.; et al. Bevacizumab and paclitaxel–carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitiveovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 779–791. [CrossRef]30. Delaloge, S.; Pérol, D.; Courtinard, C.; Brain, E.; Asselain, B.; Bachelot, T.; Debled, M.; Dieras, V.; Campone, M.; Levy, C.; et al. Paclitaxel plus bevacizumab or paclitaxel as first-line treatment for HER2-negative metastatic breast cancer in a multicenter national observational study. Ann. Oncol. 2016, 27, 1725–1732. [CrossRef] [PubMed]31. Finn, R.S.; Martin, M.; Rugo, H.S.; Jones, S.; Im, S.-A.; Gelmon, K.; Harbeck, N.; Lipatov, O.N.; Walshe, J.M.; Moulder, S.; et al. Palbociclib and Letrozole in Advanced Breast Cancer. N. Engl. J. Med. 2016, 375, 1925–1936. [CrossRef]32. Turner, N.C.; Ro, J.; André, F.; Loi, S.; Verma, S.; Iwata, H.; Harbeck, N.; Loibl, S.; Bartlett, C.H.; Zhang, K.; et al. Palbociclib in Hormone-Receptor–Positive Advanced Breast Cancer. N. Engl. J. Med. 2015, 373, 209–219. [CrossRef]33. Hortobagyi, G.N.; Stemmer, S.M.; Burris, H.A.; Yap, Y.-S.; Sonke, G.S.; Paluch-Shimon, S.; Campone, M.; Blackwell, K.L.; André, F.; Winer, E.P.; et al. Ribociclib as First-Line Therapy for HR-Positive, Advanced Breast Cancer. N. Engl. J. Med. 2016, 375, 1738–1748.[CrossRef]34. Senkus, E.; Łacko, A. Over-treatment in metastatic breast cancer. Breast 2017, 31, 309–317. [CrossRef] [PubMed]35. Cardoso, F.; Harbeck, N.; Fallowfield, L.; Kyriakides, S.; Senkus, E. ESMO Guidelines Working Group. Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2012, 23, vii11–vii19. [CrossRef] [PubMed]36. Williams, M.; Lee, L.; Werfel, T.; Joly, M.M.M.; Hicks, D.J.; Rahman, B.; Elion, D.; McKernan, C.; Sanchez, V.; Estrada, M.V.; et al. Intrinsic apoptotic pathway activation increases response to anti-estrogens in luminal breast cancers. Cell Death Dis. 2018, 9, 1–14. [CrossRef] [PubMed]37. Richman, J.; Dowsett, M. Beyond 5 years: Enduring risk of recurrence in oestrogen receptor-positive breast cancer. Nat. Rev. Clin. Oncol. 2019, 16, 296–311. [CrossRef]38. Razavi, P.; Chang, M.T.; Xu, G.; Bandlamudi, C.; Ross, D.S.; Vasan, N.; Cai, Y.; Bielski, C.M.; Donoghue, M.T.A.; Jonsson, P.; et al. The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell 2018, 34, 427–438.e6. [CrossRef]39. Boudreau, M.W.; Duraki, D.; Wang, L.; Mao, C.; Kim, J.E.; Henn, M.A.; Tang, B.; Fanning, S.W.; Kiefer, J.; Tarasow, T.M.; et al. A small-molecule activator of the unfolded protein response eradicates human breast tumors in mice. Sci. Transl. Med. 2021, 13, eabf1383. [CrossRef]40. Holland, S.J.; Pan, A.; Franci, C.; Hu, Y.; Chang, B.; Li, W.; Duan, M.; Torneros, A.; Yu, J.; Heckrodt, T.J.; et al. R428, a Selective Small Molecule Inhibitor of Axl Kinase, Blocks Tumor Spread and Prolongs Survival in Models of Metastatic Breast Cancer. Cancer Res. 2010, 70, 1544–1554. [CrossRef]41. Deng, S.; Krutilina, R.I.; Wang, Q.; Lin, Z.; Parke, D.N.; Playa, H.C.; Chen, H.; Miller, D.D.; Seagroves, T.N.; Li, W. An Orally Available Tubulin Inhibitor, VERU-111, Suppresses Triple-Negative Breast Cancer Tumor Growth and Metastasis and Bypasses Taxane Resistance. Mol. Cancer Ther. 2020, 19, 348–363. [CrossRef]42. Vernieri, C.; Corti, F.; Nichetti, F.; Ligorio, F.; Manglaviti, S.; Zattarin, E.; Rea, C.G.; Capri, G.; Bianchi, G.V.; De Braud, F. Everolimus versus alpelisib in advanced hormone receptor-positive HER2-negative breast cancer: Targeting different nodes of the PI3K/AKT/mTORC1 pathway with different clinical implications. Breast Cancer Res. 2020, 22, 1–13. [CrossRef]43. Jager, A.; De Vries, E.G.E.; Oordt, C.W.M.-V.D.H.V.; Neven, P.; Venema, C.M.; Glaudemans, A.W.J.M.; Wang, Y.; Bagley, R.G.; Conlan, M.G.; Aftimos, P. A phase 1b study evaluating the effect of elacestrant treatment on estrogen receptor availability and estradiol binding to the estrogen receptor in metastatic breast cancer lesions using 18F-FES PET/CT imaging. Breast Cancer Res. 2020, 22, 1–11. [CrossRef]44. Liang, J.; Zbieg, J.R.; Blake, R.A.; Chang, J.H.; Daly, S.; DiPasquale, A.G.; Friedman, L.S.; Gelzleichter, T.; Gill, M.; Giltnane, J.M.; et al. GDC-9545 (Giredestrant): A Potent and Orally Bioavailable Selective Estrogen Receptor Antagonist and Degrader with an Exceptional Preclinical Profile for ER+ Breast Cancer. J. Med. Chem. 2021, 64, 11841–11856. [CrossRef] [PubMed]45. Scott, J.S.; Moss, T.A.; Balazs, A.; Barlaam, B.; Breed, J.; Carbajo, R.J.; Chiarparin, E.; Davey, P.R.J.; Delpuech, O.; Fawell, S.; et al. Discovery of AZD9833, a Potent and Orally Bioavailable Selective Estrogen Receptor Degrader and Antagonist. J. Med. Chem. 2020, 63, 14530–14559. [CrossRef] [PubMed]46. Doll, R.; Peto, R. The Causes of Cancer: Quantitative Estimates of Avoidable Risks of Cancer in the United States Today. J. Natl. Cancer Inst. 1981, 66, 1192–1308. [CrossRef]47. Nicodemus, K.K.; Jacobsr, D.R., Jr.; Folsom, A.R. Whole and refined grain intake and risk of incident postmenopausal breast cancer (United States). Cancer Causes Control 2001, 12, 917–925. [CrossRef] [PubMed]48. Dydjow-Bendek, D.; Zagozdzon, P. Total Dietary Fats, Fatty Acids, and Omega-3/Omega-6 Ratio as Risk Factors of Breast Cancer in the Polish Population-a Case-Control Study. In Vivo 2020, 34, 423–431. [CrossRef]49. Tsilidis, K.K.; Travis, R.C.; Appleby, P.N.; Allen, N.E.; Lindström, S.; Albanes, D.; Ziegler, R.G.; McCullough, M.L.; Siddiq, A.; Barricarte, A.; et al. Insulin-like growth factor pathway genes and blood concentrations, dietary protein and risk of prostate cancer in the NCI Breast and Prostate Cancer Cohort Consortium (BPC3). Int. J. Cancer 2013, 133, 495–504. [CrossRef]50. Williams, C.M.; Dickerson, J.W. Dietary fat, hormones and breast cancer: The cell membrane as a possible site of interaction of these two risk factors. Eur. J. Surg. Oncol. 1987, 13, 89–104.51. Levi, F.; La Vecchia, C.; Gulie, C.; Negri, E. Dietary Factors and Breast-Cancer Risk in Vaud, Switzerland. Nutr. Cancer Int. J. 1993, 19, 327–335. [CrossRef]52. Mauvais-Jarvis, F.; Clegg, D.J.; Hevener, A.L. The Role of Estrogens in Control of Energy Balance and Glucose Homeostasis. Endocr. Rev. 2013, 34, 309–338. [CrossRef]53. Madak-Erdogan, Z.; Band, S.; Zhao, Y.C.; Smith, B.P.; Kulkoyluoglu-Cotul, E.; Zuo, Q.; Casiano, A.S.; Wrobel, K.; Rossi, G.; Smith, R.L.; et al. Free fatty acids rewire cancer metabolism in obesity-associated breast cancer via estrogen receptor and mTOR signaling. Cancer Res. 2019, 79, 2494–2510. [CrossRef]54. Zuo, Q.; Band, S.; Kesavadas, M.; Erdogan, Z.M. Obesity and Postmenopausal Hormone Receptor-positive Breast Cancer: Epidemiology and Mechanisms. Endocrinology 2021, 162, bqab195. [CrossRef] [PubMed]55. Jiralerspong, S.; Goodwin, P.J. Obesity and Breast Cancer Prognosis: Evidence, Challenges, and Opportunities. J. Clin. Oncol. 2016, 34, 4203–4216. [CrossRef] [PubMed]56. Suzuki, R.; Orsini, N.; Saji, S.; Key, T.J.; Wolk, A. Body weight and incidence of breast cancer defined by estrogen and progesterone receptor status-A meta-analysis. Int. J. Cancer 2009, 124, 698–712. [CrossRef] [PubMed]57. Iyengar, N.M.; Arthur, R.; Manson, J.E.; Chlebowski, R.T.; Kroenke, C.H.; Peterson, L.; Cheng, T.D.; Feliciano, E.; Lane, D.; Luo, J.; et al. Association of Body Fat and Risk of Breast Cancer in Postmenopausal Women with Normal Body Mass Index A Secondary Analysis of a Randomized Clinical Trial and Observational Study. JAMA Oncol. 2019, 5, 155–163. [CrossRef]58. Picon-Ruiz, M.; Morata-Tarifa, C.; Valle-Goffin, J.J.; Friedman, E.R.; Slingerland, J.M. Obesity and adverse breast cancer risk and outcome: Mechanistic insights and strategies for intervention. CA Cancer J. Clin. 2017, 67, 378–397. [CrossRef]59. Keum, N.; Greenwood, D.C.; Lee, D.H.; Kim, R.; Aune, D.; Ju, W.; Hu, F.B.; Giovannucci, E.L. Adult Weight Gain and Adiposity- Related Cancers: A Dose-Response Meta-Analysis of Prospective Observational Studies. J. Natl. Cancer Inst. 2015, 107, djv088.60. Renehan, A.G.; Tyson, M.; Egger, M.; Heller, R.F.; Zwahlen, M. Body-mass index and incidence of cancer: A systematic review and meta-analysis of prospective observational studies. Lancet 2008, 371, 569–578. [CrossRef]61. Chan, D.S.M.; Vieira, A.R.; Aune, D.; Bandera, E.V.; Greenwood, D.C.; McTiernan, A.; Rosenblatt, D.N.; Thune, I.; Vieira, R.; Norat, T. Body mass index and survival in women with breast cancer—systematic literature review and meta-analysis of 82 follow-up studies. Ann. Oncol. 2014, 25, 1901–1914. [CrossRef]62. Gathirua-Mwangi, W.G.; Palmer, J.R.; Champion, V.; Castro-Webb, N.; Stokes, A.C.; Adams-Campbell, L.; Marley, A.R.; Forman, M.R.; Rosenberg, L.; Bertrand, K.A. Maximum and Time-Dependent Body Mass Index and Breast Cancer Incidence Among Postmenopausal Women in the Black Women’s Health Study. Am. J. Epidemiol. 2022, 191, 646–654. [CrossRef]63. Chauhan, R.; Trivedi, V.; Rani, R.; Singh, U. A comparative analysis of body mass index with estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 status in pre- and postmenopausal breast cancer patients. J. Mid-Life Health 2020, 11, 210–216. [CrossRef]64. Statovci, D.; Aguilera, M.; Mac Sharry, J.; Melgar, S. The Impact of Western Diet and Nutrients on the Microbiota and Immune Response at Mucosal Interfaces. Front. Immunol. 2017, 8, 838. [CrossRef] [PubMed]65. Garcia-Montero, C.; Fraile-Martínez, O.; Gómez-Lahoz, A.M.; Pekarek, L.; Castellanos, A.J.; Noguerales-Fraguas, F.; Coca, S.; Guijarro, L.G.; García-Honduvilla, N.; Asúnsolo, A.; et al. Nutritional Components in Western Diet Versus Mediterranean Diet at the Gut Microbiota-Immune System Interplay. Implications for Health and Disease. Nutrients 2021, 13, 699. [CrossRef] [PubMed]66. Zinöcker, M.K.; Lindseth, I.A. The Western Diet–Microbiome-Host Interaction and Its Role in Metabolic Disease. Nutrients 2018, 10, 365. [CrossRef] [PubMed]67. Cully, M.; You, H.; Levine, A.J.; Mak, T.W. Beyond PTEN mutations: The PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat. Rev. Cancer 2006, 6, 184–192. [CrossRef] [PubMed]68. Haythorne, E.; Rohm, M.; Van De Bunt, M.; Brereton, M.F.; Tarasov, A.I.; Blacker, T.S.; Sachse, G.; Dos Santos, M.S.; Exposito, R.T.; Davis, S.; et al. Diabetes causes marked inhibition of mitochondrial metabolism in pancreatic β-cells. Nat. Commun. 2019, 10, 1–17. [CrossRef]69. Newsholme, P.; Keane, K.N.; Carlessi, R.; Cruzat, V. Oxidative stress pathways in pancreatic β-cells and insulin-sensitive cells and tissues: Importance to cell metabolism, function, and dysfunction. Am. J. Physiol. Cell Physiol. 2019, 317, C420–C433. [CrossRef]70. Klement, R.J.; Kammerer, U. Is there a role for carbohydrate restriction in the treatment and prevention of cancer? Nutr. Metab. 2011, 8, 1–16. [CrossRef]71. LaPensee, C.R.; Hugo, E.R.; Ben-Jonathan, N. Insulin Stimulates Interleukin-6 Expression and Release in LS14 Human Adipocytes through Multiple Signaling Pathways. Endocrinology 2008, 149, 5415–5422. [CrossRef]72. Makino, T.; Noguchi, Y.; Yoshikawa, T.; Doi, C.; Nomura, K. Circulating interleukin 6 concentrations and insulin resistance in patients with cancer. Br. J. Surg. 1998, 85, 1658–1662. [CrossRef]73. Mccall, J.L.; Tuckey, J.A.; Parry, B.R. Serum Tumor-Necrosis-Factor-Alpha and Insulin Resistance in Gastrointestinal Cancer. Br. J. Surg. 1992, 79, 1361–1363. [CrossRef]74. Tian, M.; Zhang, H.; Nakasone, Y.; Mogi, K.; Endo, K. Expression of Glut-1 and Glut-3 in untreated oral squamous cell carcinoma compared with FDG accumulation in a PET study. Eur. J. Nucl. Med. Mol. Imaging 2004, 31, 5–12. [CrossRef] [PubMed]75. Zhang, H.-L.; Wang, M.-D.; Zhou, X.; Qin, C.-J.; Fu, G.-B.; Tang, L.; Wu, H.; Huang, S.; Zhao, L.-H.; Zeng, M.; et al. Blocking preferential glucose uptake sensitizes liver tumor-initiating cells to glucose restriction and sorafenib treatment. Cancer Lett. 2017, 388, 1–11. [CrossRef] [PubMed]76. Maldonado, R.; Talana, C.A.; Song, C.; Dixon, A.; Uehara, K.; Weichhaus, M. β-hydroxybutyrate does not alter the effects of glucose deprivation on breast cancer cells. Oncol. Lett. 2021, 21, 1. [CrossRef] [PubMed]77. Varghese, S.; Samuel, S.M.; Varghese, E.; Kubatka, P.; Büsselberg, D. High Glucose Represses the Anti-Proliferative and Pro- Apoptotic Effect of Metformin in Triple Negative Breast Cancer Cells. Biomolecules 2019, 9, 16. [CrossRef]78. Zuo, Q.; Mogol, A.N.; Liu, Y.-J.; Casiano, A.S.; Chien, C.; Drnevich, J.; Imir, O.B.; Kulkoyluoglu-Cotul, E.; Park, N.H.; Shapiro, D.J.; et al. Targeting metabolic adaptations in the breast cancer-liver metastatic niche using dietary approaches to improve endocrine therapy efficacy. Mol. Cancer Res. 2022, 20, 923–937. [CrossRef]79. Healy, M.E.; Chow, J.D.; Byrne, F.L.; Breen, D.S.; Leitinger, N.; Li, C.; Lackner, C.; Caldwell, S.H.; Hoehn, K.L. Dietary effects on liver tumor burden in mice treated with the hepatocellular carcinogen diethylnitrosamine. J. Hepatol. 2015, 62, 599–606. [CrossRef]80. Bechmann, L.P.; Hannivoort, R.A.; Gerken, G.; Hotamisligil, G.S.; Trauner, M.; Canbay, A. The interaction of hepatic lipid and glucose metabolism in liver diseases. J. Hepatol. 2012, 56, 952–964. [CrossRef]81. Zhong, X.-Y.; Yuan, X.-M.; Xu, Y.-Y.; Yin, M.; Yan, W.-W.; Zou, S.-W.; Wei, L.-M.; Lu, H.-J.; Wang, Y.-P.; Lei, Q.-Y. CARM1 Methylates GAPDH to Regulate Glucose Metabolism and Is Suppressed in Liver Cancer. Cell Rep. 2018, 24, 3207–3223. [CrossRef]82. Krstic, J.; Reinisch, I.; Schindlmaier, K.; Galhuber, M.; Riahi, Z.; Berger, N.; Kupper, N.; Moyschewitz, E.; Auer, M.; Michenthaler, H.; et al. Fasting improves therapeutic response in hepatocellular carcinoma through p53-dependent metabolic synergism. Sci. Adv. 2022, 8, eabh2635. [CrossRef]83. Wahdan-Alaswad, R.S.; Edgerton, S.M.; Salem, H.S.; Thor, A.D. Metformin Targets Glucose Metabolism in Triple Negative Breast Cancer. J. Oncol. Transl. Res. 2018, 4, 129. [CrossRef]84. Roy, R.; Hahm, E.R.; White, A.G.; Anderson, C.J.; Singh, S.V. AKT-dependent sugar addiction by benzyl isothiocyanate in breast cancer cells. Mol. Carcinog. 2019, 58, 996–1007. [CrossRef] [PubMed]85. Gluschnaider, U.; Hertz, R.; Ohayon, S.; Smeir, E.; Smets, M.; Pikarsky, E.; Bar-Tana, J. Long-Chain Fatty Acid Analogues Suppress Breast Tumorigenesis and Progression. Cancer Res. 2014, 74, 6991–7002. [CrossRef] [PubMed]86. Wei, M.; Brandhorst, S.; Shelehchi, M.; Mirzaei, H.; Cheng, C.W.; Budniak, J.; Groshen, S.; Mack, W.J.; Guen, E.; Di Biase, S.; et al. Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci. Transl. Med. 2017, 9. [CrossRef] [PubMed]87. Poff, A.; Ari, C.; Arnold, P.; Seyfried, T.; D’Agostino, D. Ketone supplementation decreases tumor cell viability and prolongs survival of mice with metastatic cancer. Int. J. Cancer 2014, 135, 1711–1720. [CrossRef] [PubMed]88. Allen, B.G.; Bhatia, S.K.; Anderson, C.M.; Eichenberger-Gilmore, J.M.; Sibenaller, Z.A.; Mapuskar, K.A.; Schoenfeld, J.D.; Buatti, J.M.; Spitz, D.R.; Fath, M.A. Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biol. 2014, 2, 963–970. [CrossRef]89. Rigo, P.; Paulus, P.; Kaschten, B.J.; Hustinx, R.; Bury, T.; Jerusalem, G.; Benoit, T.; Foidart-Willems, J. Oncological applications ofpositron emission tomography with fluorine-18 fluorodeoxyglucose. Eur. J. Nucl. Med. 1996, 23, 1641–1674. [CrossRef]90. Kaji, K.; Nishimura, N.; Seki, K.; Sato, S.; Saikawa, S.; Nakanishi, K.; Furukawa, M.; Kawaratani, H.; Kitade, M.; Moriya, K.; et al. Sodium glucose cotransporter 2 inhibitor canagliflozin attenuates liver cancer cell growth and angiogenic activity by inhibiting glucose uptake. Int. J. Cancer 2018, 142, 1712–1722. [CrossRef]91. Zhang, X.; Qiao, Y.; Wu, Q.; Chen, Y.; Zou, S.; Liu, X.; Zhu, G.; Zhao, Y.; Chen, Y.; Yu, Y.; et al. The essential role of YAP O-GlcNAcylation in high-glucose-stimulated liver tumorigenesis. Nat. Commun. 2017, 8, 1–15. [CrossRef]92. Bu, P.; Chen, K.-Y.; Xiang, K.; Johnson, C.; Crown, S.B.; Rakhilin, N.; Ai, Y.; Wang, L.; Xi, R.; Astapova, I.; et al. Aldolase B-Mediated Fructose Metabolism Drives Metabolic Reprogramming of Colon Cancer Liver Metastasis. Cell Metab. 2018, 27, 1249–1262.e4. [CrossRef]93. Warburg, O. On the Origin of Cancer Cells. Science 1956, 123, 309–314. [CrossRef]94. Anykin-Burns, N.; Ahmad, I.A.; Zhu, Y.; Oberley, L.W.; Spitz, D.R. Increased levels of superoxide and hydrogen peroxide mediate the differential susceptibility of cancer cells vs. Normal cells to glucose deprivation. Biochem. J. 2009, 418, 29–37. [CrossRef] [PubMed]95. Boros, L.G.; Lee, P.W.N.; Brandesa, J.L.; Cascante, M.; Muscarellaa, P.; Schirmera, W.J.; Melvina, W.S.; Ellisona, E.C. Nonoxidative pentose phosphate pathways and their direct role in ribose synthesis in tumors: Is cancer a disease of cellular glucose metabolism? Med. Hypotheses 1998, 50, 55–59.[CrossRef]96. Weber, D.D.; Aminazdeh-Gohari, S.; Kofler, B. Ketogenic diet in cancer therapy. Aging 2018, 10, 164–165. [CrossRef] [PubMed]97. Wallace, D.C. Mitochondria and cancer. Nat. Rev. Cancer 2012, 12, 685–698. [CrossRef] [PubMed]98. Buettner, G.R. Superoxide dismutase in redox biology: The roles of superoxide and hydrogen peroxide. Anti-Cancer Agents Med. Chem. 2011, 11, 341–346. [CrossRef] [PubMed]99. Veech, R.L. Ketone ester effects on metabolism and transcription. J. Lipid Res. 2014, 55, 2004–2006. [CrossRef]100. Hopkins, B.D.; Pauli, C.; Du, X.; Wang, D.G.; Li, X.; Wu, D.; Amadiume, S.C.; Goncalves, M.D.; Hodakoski, C.; Lundquist, M.R.; et al. Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 2018, 560, 499–503, Erratum in Nature 2018, 563, E24. [CrossRef]101. Ho, V.W.; Leung, K.; Hsu, A.; Luk, B.; Lai, J.; Shen, S.Y.; Minchinton, A.I.; Waterhouse, D.; Bally, M.; Lin, W.; et al. A Low Carbohydrate, High Protein Diet Slows Tumor Growth and Prevents Cancer Initiation. Cancer Res. 2011, 71, 4484–4493. [CrossRef]102. Vanderheiden, M.G.; Cantley, L.C.; Thompson, C.B. UnderstandingtheWarburgEffect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [CrossRef]103. Byrne, F.L.;Hargett, S.R.; Lahiri, S.; Roy, R.J.; Berr, S. S.; Caldwell, S.H.; Hoehn, K.L. Serial MRI Imaging Reveals Minimal Impact of Ketogenic Diet on Established Liver Tumor Growth. Cancers 2018, 10, 312. [CrossRef]104. Tan-Shalaby, J. Ketogenic diets, and cancer: Emerging evidence.Fed.Pract.2017,34,37S.[PubMed]105. Allen, B.G.; Bhatia, S.K.; Buatti, J.; Brandt, K.E.; Lindholm, K.E.; Button, A.; Szweda, L.I.; Smith, B.J.; Spitz, D.R.; Fath, M.A. Ketogenic Diets Enhance Oxidative Stress and Radio-Chemo-Therapy Responses in Lung Cancer Xenografts. Clin. Cancer Res. 2013, 19, 3905–3913. [CrossRef] [PubMed]106.Abdelwahab, M.G.; Fenton, K.E.; Preul, M.C.; Rho, J.M.; Lynch,A.; Stafford, P.; Scheck, A.C. The Ketogenic Diet Isan Effective Adjuvant to Radiation Therapy for the Treatment of Malignant Glioma. PLoS ONE 2012, 7, e36197. [CrossRef] [PubMed]107. Fokidis, H.B.;Chin, M.Y.;Ho, V.W.;Adomat, H.H.; Soma,K.K.; Fazli,L.; Nip, K.M.; Cox, M.; Krystal, G.; Zoubeidi, A.; et. A low carbohydrate, high protein diet suppresses intratumoral androgen synthesis and slows castration-resistant prostate tumorgrowth in mice. J. Steroid Biochem. Mol. Biol. 2015, 150, 35–45. [CrossRef] [PubMed]108. Ho, V.W.; Hamilton, M.J.; Dang, N.-H.T.; Hsu, B.E.; Adomat, H.H.;Guns, E.S.; Weljie,A.; Samudio, I.; Bennewith, K.L.; Krystal, G. A low carbohydrate, high protein diet combined with celecoxib markedly reduces metastasis. Carcinogenesis 2014, 35, 2291–2299.[CrossRef] [PubMed]109. Martuscello,R.T.; Vedam-Mai,V.; McCarthy, D.J.; Schmoll, M.E.; Jundi, M.A.; Louviere, C.D.; Griffith, B.G.; Skinner, C.L.; Suslov, O.; Deleyrolle, L.P.; et al. A Supplemented High-Fat Low-Carbohydrate Diet for the Treatment of Glioblastoma. Clin. Cancer Res. 2016, 22, 2482–2495. [CrossRef]110. Caffa,I.; Spagnolo,V.; Vernieri,C.; Valdemarin,F.; Becherini,P.; Wei,M.; Brandhorst,S.; Zucal,C.; Driehuis,E.; Ferrando,L. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 2020, 583, 620–624. [CrossRef]111. Klement,R.J.;Champ,C.E.;Otto,C.;Kämmerer,U.Anti-TumorEffectsofKetogenicDietsinMice:AMeta-Analysis.PLoSONE 2016, 11, e0155050. [CrossRef]112. Yang, L.; TeSlaa, T.; Ng, S.; Nofal, M.; Wang, L.; Lan, T.; Zeng, X.; Cowan, A.; McBride, M.; Lu, W.; et al. Ketogenic diet and chemotherapy combine to disrupt pancreatic cancer metabolism and growth. Med 2022, 3, 119–136.e8. [CrossRef] [PubMed]113. DiBiase,S.; Shim,H.S.;Kim,K.H.; Vinciguerra,M.; Rappa,F.; Wei,M.; Brandhorst,S.; Cappello,F.; Mirzaei,H.; Lee,C.;etal. Fasting regulates EGR1 and protects from glucose-and dexamethasone-dependent sensitization to chemotherapy. PLoS Biol. 2017, 15, e2001951. [CrossRef]114. Salvadori,G.; Zanardi,F.; Iannelli,F.; Lobefaro,R.; Vernieri,C.;Longo,V.D.Fasting-mimicking diet blocks triple-negative breast cancer and cancer stem cell escape. Cell Metab. 2021, 33, 2247–2259.e6. [CrossRef] [PubMed]115. Elgendy,M.; Cirò,M.; Hosseini,A.; Weiszmann,J.; Mazzarella,L.;Ferrari,E.; Cazzoli,R.; Curigliano,G.; DeCensi,A.; Bonanni, B.; et al. Combination of Hypoglycemia and Metformin Impairs Tumor Metabolic Plasticity and Growth by Modulating the PP2A-GSK3β-MCL-1 Axis. Cancer Cell 2019, 35, 798–815.e5. [CrossRef] [PubMed]116. Puchalska,P.; Crawford,P.A. Multidimensional Rolesof Ketone Bodies in Fuel Metabolism, Signaling,and Therapeutics. Cell Metab. 2017, 25, 262–284. [CrossRef] [PubMed]117. Laffel,L. Ketone bodies: Are view of physiology, pathophysiology and application of monitoring to diabetes. Diabetes/Metab.Res. Rev. 1999, 15, 412–426. [CrossRef]118. Newman,J.C.; Verdin,E. β-Hydroxybutyrate: A Signaling Metabolite. Annu.Rev.Nutr.2017,37,51–76.[CrossRef]119. Dhillon, K.K.; Gupta, S.Biochemistry, Ketogenesis. InStat Pearls; Stat Pearls Publishing: TreasureIsland, FL, USA,2022.Available online: https://www.ncbi.nlm.nih.gov/books/NBK493179/ (accessed on 10 February 2022).120. Kim,D.Y.;Rho,J.M.Theketogenicdietandepilepsy.Curr.Opin.Clin.Nutr.Metab.Care2008,11,113–120.[CrossRef]121. Duan,Y.; Zhang,Y.; Dong,H.; Wang,Y.;Zhang,J. Effects of dietarypoly-beta-hydroxybutyrate(PHB) on microbiotacomposition and the mTOR signaling pathway in the intestines of litopenaeus vannamei. J. Microbiol. 2017, 55, 946–954. [CrossRef]122. Huang,C.; Wang,P.; Xu,X.; Zhang,Y.; Gong,Y.; Hu,W.; Gao,M.; Wu,Y.; Ling,Y.; Zhao,X.; etal.Theketonebodymetabolite beta-hydroxybutyrate induces an antidepression-associated ramification of microglia via HDACs inhibition-triggered Akt-small RhoGTPase activation. Glia 2018, 66, 256–278. [CrossRef]123. Vernieri, C.; Fucà, G.; Ligorio, F.; Huber, V.; Vingiani, A.; Iannelli, F.; Raimondi, A.; Rinchai, D.; Frigè, G.; Belfiore, A.; et al. Fasting-Mimicking Diet Is Safe and Reshapes Metabolism and Antitumor Immunity in Patients with Cancer. Cancer Discov. 2022, 12, 90–107. [CrossRef]124. Han, Y.-M.; Ramprasath, T.; Zou, M.-H. β-hydroxybutyrate and its metabolic effects on age-associated pathology.Exp.Mol.Med. 2020, 52, 548–555. [CrossRef]125. Kim, D.H.; Park, M.H.; Ha, S.; Bang, E.J.; Lee, Y.; Lee, A.K.; Lee, J.; Yu, B.P.; Chung, H.Y. Anti-inflammatory action of beta-hydroxybutyrate via modulation of PGC-1alpha and FoxO1, mimicking calorie restriction. Aging 2019, 11, 1283–1304. [CrossRef]126. Shimazu,T.; Hirschey,M.D.; Newman,J.; He,W.; Shirakawa,K.; LeMoan,N.; Grueter,C.A.; Lim,H.; Saunders,L.R.; Stevens, R.D.; et al. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 2013, 339, 211–214. [CrossRef] [PubMed]127. Pant,K.; Peixoto,E.; Richard,S.; Gradilone,S.A. Role of Histone Deacetylases in Carcinogenesis: Potential Role in Cholangiocarcinoma. Cells 2020, 9, 780. [CrossRef]128. Martinez-Outschoorn, U.E.; Prisco, M.; Ertel, A.; Tsirigos, A.; Lin, Z.; Pavlides, S.; Wang, C.; Flomenberg, N.; Knudsen, E.S.; Howell, A. Ketones and lactate increase cancer cell “stemness,” driving recurrence, metastasis and poor clinical outcome in breast cancer: Achieving personalized medicine via Metabolo-Genomics. Cell Cycle 2011, 10, 1271–1286. [CrossRef] [PubMed]129. Bonuccelli,G.; Tsirigos,A.; Whitaker-Menezes,D.; Pavlides,S.; Pestell,R.G.; Chiavarina,B.; Frank,P.G.; Flomenberg,N.; Howell, A.; Martinez-Outschoorn, U.E.; et al. Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle 2010, 9, 3506–3514. [CrossRef] [PubMed]130. Huang,C.; Chang,P.; Kuo,W.; Chen,C.; Jeng,Y.; Chang,K.; Shew,J.; Hu,C.; Lee,W. Adipocytes promote malignant growth of breast tumours with monocarboxylate transporter 2 expression via beta-hydroxybutyrate. Nat. Commun. 2017, 8, 14706. [CrossRef] [PubMed]131. Rodrigues, L.M.; Uribe-Lewis, S.; Madhu, B.; Honess, D.J.; Stubbs, M.; Griffiths, J.R. The action of β-hydroxybutyrate on the growth, metabolism and global histone H3 acetylation of spontaneous mouse mammary tumours: Evidence of a β- hydroxybutyrate paradox. Cancer Metab. 2017, 5, 1–13. [CrossRef]132. Shukla,S.K.; Gebregiworgis,T.; Purohit,V.; Chaika,N.V.; Gunda,V.;Radhakrishnan,P.; Mehla,K.; Pipinos,I.I.;Powers,R.; Yu, F.; et al. Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia. Cancer Metab. 2014, 2, 18. [CrossRef]133. Mikami, D.; Kobayashi, M.; Uwada, J.; Yazawa, T.; Kamiyama, K.; Nishimori, K.; Nishikawa, Y.; Nishikawa, S.; Yokoi, S.; Taniguchi, T.; et al. β-Hydroxybutyrate enhances the cytotoxic effect of cisplatin via the inhibition of HDAC/survivin axis in human hepatocellular carcinoma cells. J. Pharmacol. Sci. 2020, 142, 1–8. [CrossRef]134. Schmidt,M.; Pfetzer,N.; Schwab,M.; Strauss,I.; Kämmerer,U. Effects of a ketogenic diet on the quality of life in 16 patients with advanced cancer: A pilot trial. Nutr. Metab. 2011, 8, 54. [CrossRef]135. Bartmann,C.; Raman,S.R.J.; Flöter,J.; Schulze,A.; Bahlke,K.; Willingstorfer,J.; Strunz,M.; Wöckel,A.; Klement,R.J.; Kapp, M.; et al. Beta-hydroxybutyrate (3-OHB) can influence the energetic phenotype of breast cancer cells, but does not impact their proliferation and the response to chemotherapy or radiation. Cancer Metab. 2018, 6, 8. [CrossRef] [PubMed]136. Donohoe,D.R.; Collins,L.B.; Wali,A.; Bigler,R.; Sun,W.; Bultman,S.J. The Warburg Effect Dictates the Mechanism of Butyrate-Mediated Histone Acetylation and Cell Proliferation. Mol. Cell 2012, 48, 612–626. [CrossRef] [PubMed]137. Lupton,J.R. Microbial Degradation Products Influence Colon Cancer Risk: The Butyrate Controversy.J.Nutr.2004,134,479–482. [CrossRef]138. Luo,S.; Li,Z.; Mao,L.; Chen,S.; Sun,S. Sodium butyrate induces autophagy incolorectal cancer cells through LKB1/AMPK signaling. J. Physiol. Biochem. 2019, 75, 53–63. [CrossRef] [PubMed]139. Salimi,V.; Shahsavari,Z.; Safizadeh,B.; Hosseini,A.;Khademian,N.;Tavakoli-Yaraki,M. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis. 2017, 16, 1–11. [CrossRef] [PubMed]140. Vernieri, C.; Ligorio, F.; Zattarin, E.; Rivoltini, L.; De Braud, F. Fasting-mimicking diet plus chemotherapy in breast cancer treatment. Nat. Commun. 2020, 11, 1–4. [CrossRef]141. DeGroot,S.; Lugtenberg,R.T.; Cohen,D.; Welters,M.J.P.; Ehsan,I.; Vreeswijk,M.P.G.; Smit,V.T.H.B.M.; deGraaf,H.; Heijns,J.B.; Portielje, J.E.A.; et al. Fasting mimicking diet as an adjunct to neoadjuvant chemotherapy for breast cancer in the multicentre randomized phase 2 DIRECT trial. Nat. Commun. 2020, 11, 1–9. [CrossRef]142. DiBiase,S.; Lee,C.; Brandhorst,S.; Manes,B.; Buono,R.; Cheng,C.-W.; Cacciottolo,M.; Martin-Montalvo,A.; DeCabo,R.; Wei, M.; et al. Fasting-Mimicking Diet Reduces HO-1 to Promote T Cell-Mediated Tumor Cytotoxicity. Cancer Cell 2016, 30, 136–146. [CrossRef]
More
This entry is offline, you can click here to edit this entry!
Video Production Service