Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade

Gladys Ferrere, Maryam Tidjani Alou, Peng Liu, Anne-Gaëlle Goubet, Marine Fidelle, Oliver Kepp, Sylvère Durand, Valerio Iebba, Aurélie Fluckiger, Romain Daillère, Cassandra Thelemaque, Claudia Grajeda-Iglesias, Carolina Alves Costa Silva, Fanny Aprahamian, Déborah Lefevre, Liwei Zhao, Bernhard Ryffel, Emeline Colomba, Monica Arnedos, Damien Drubay, Conrad Rauber, Didier Raoult, Francesco Asnicar, Tim Spector, Nicola Segata, Lisa Derosa, Guido Kroemer, Laurence Zitvogel, Gladys Ferrere, Maryam Tidjani Alou, Peng Liu, Anne-Gaëlle Goubet, Marine Fidelle, Oliver Kepp, Sylvère Durand, Valerio Iebba, Aurélie Fluckiger, Romain Daillère, Cassandra Thelemaque, Claudia Grajeda-Iglesias, Carolina Alves Costa Silva, Fanny Aprahamian, Déborah Lefevre, Liwei Zhao, Bernhard Ryffel, Emeline Colomba, Monica Arnedos, Damien Drubay, Conrad Rauber, Didier Raoult, Francesco Asnicar, Tim Spector, Nicola Segata, Lisa Derosa, Guido Kroemer, Laurence Zitvogel

Abstract

Limited experimental evidence bridges nutrition and cancer immunosurveillance. Here, we show that ketogenic diet (KD) - or its principal ketone body, 3-hydroxybutyrate (3HB), most specifically in intermittent scheduling - induced T cell-dependent tumor growth retardation of aggressive tumor models. In conditions in which anti-PD-1 alone or in combination with anti-CTLA-4 failed to reduce tumor growth in mice receiving a standard diet, KD, or oral supplementation of 3HB reestablished therapeutic responses. Supplementation of KD with sucrose (which breaks ketogenesis, abolishing 3HB production) or with a pharmacological antagonist of the 3HB receptor GPR109A abolished the antitumor effects. Mechanistically, 3HB prevented the immune checkpoint blockade-linked upregulation of PD-L1 on myeloid cells, while favoring the expansion of CXCR3+ T cells. KD induced compositional changes of the gut microbiota, with distinct species such as Eisenbergiella massiliensis commonly emerging in mice and humans subjected to carbohydrate-low diet interventions and highly correlating with serum concentrations of 3HB. Altogether, these results demonstrate that KD induces a 3HB-mediated antineoplastic effect that relies on T cell-mediated cancer immunosurveillance.

Keywords: Cancer; Immunotherapy; Metabolism; Mouse models; Oncology.

Conflict of interest statement

Conflict of interest: LZ, RD, and GK are founders of EverImmune.

Figures

Figure 1. Ketogenic diet decreases melanoma and…
Figure 1. Ketogenic diet decreases melanoma and renal cell tumor growth.
(A–B) Growth kinetics of RET melanoma in C57BL/6J mice. The minimal tumorigenic dose of RET melanoma cells was inoculated s.c. the day of starting the dietary intervention. Normal diet (ND) versus ketogenic diet (KD). Tumor size was monitored 3 times a week for 12 days. Each curve represents the tumor kinetics of 1 animal (A), and the mean ± SEM of tumor size was calculated for both nutritional interventions (B). The graphs depict the concatenation of 5 independent experiments involving 5–6 mice/group (n = 29–30 mice). (C) Monitoring of RENCA progression using bioluminescence imaging of luciferase activity in 4 representative mice among 15 BALB/c mice fed ND versus KD. (D) The ratio of luminescence at day 15 (D15) versus day 7 after orthotopic tumor injection was calculated for each diet (D15, IVIS measurement; D7, day of randomization). All experiments were composed of 5–7 mice/group and were performed at least twice, yielding similar results (n = 14 for ND and n = 12 for KD). Dedicated software (https://kroemerlab.shinyapps.io/TumGrowth/) (A and B) and Student’s t test. *P < 0.05 (D).
Figure 2. Ketone bodies accumulate in tissues…
Figure 2. Ketone bodies accumulate in tissues of ketogenic diet fed mice.
(A) Follow up of mouse weight over time until sacrifice in tumor bearers fed normal diet (ND) versus ketogenic diet (KD) (n = 30 for each groups). (B and C) Heatmap of the nonsupervised hierarchical clustering highlighting differences in the metabolic profiling of C57BL/6J mice fed ND (n = 20) versus KD (n = 21) in plasma (B) and PCA based on Bray-Curtis Dissimilarity Index showing significant compositional differences across diet types (C) (PERMANOVA, P = 0.001). Arrows indicate 3-hydroxybutyrate (3HB) and acetoacetate positions in the heatmap. (D and E) Concentrations of acetoacetate and 3HB in 3 compartments of tumor bearers. A representative experiment involving 5 mice/group is depicted out of 2 metabolomics profiles yielding similar results (n = 3 in ND and n = 5 in KD). (F) Spearman correlations between levels of 3HB in plasma and RET tumor size at day 12 indicating r and P values for the KD group. Not significant for the ND group. ANOVA (A) and Student’s t test statistical analyses of mean ± SEM. *P < 0.05 (D and E).
Figure 3. Ketone body 3-hydroxybutyrate (3HB) is…
Figure 3. Ketone body 3-hydroxybutyrate (3HB) is necessary and sufficient to account for the anticancer effects of ketogenic diet.
(A and B) Similar experimental setting as in Figure 1, A and B, comparing RET tumor size at day 12 of each nutritional intervention (normal diet [ND], ketogenic diet [KD], 3-hydroxybutyrate per os [3HBpo] versus 3-hydroxybutyrate i.p. [3HBip]) for all 12–15 mouse tumors from 3 independent experiments (A) and the complete regression rates for all the experiments performed (n, number of independent experiments encompassing 5–6 mice/group, SEM of percentage of tumor free mice across 3–7 experiments) (B). (C) Pharmacological inhibition of GPR109A (versus PBS as control) using i.p. daily administration of mepenzolate bromide (C21H26BrNO3). Tumor sizes at day 12 are depicted for each group. Data from 2–3 experiments involving 5–6 mice/group are depicted (n = 20 for ND, KD, and 3HBpo without mepenzolate and n = 24 for ND, KD, 3HBpo + mepenzolate. (DF) Effects of sucrose supplementation on the antitumor (D) and metabolic effects (E and F) mediated by KD. Id. as in Figure 1, A and B, showing RET tumor sizes at day 12 after various dietary interventions (D) and the 2 ketone body (KB) plasma metabolites as in Figure 2, D and E (E and F). Results from 1 representative experiment out of 2 are depicted (n = 30 for group without sucrose and n = 12 with sucrose groups). Global comparison using Kruskall-Wallis test, with a post hoc multiple comparison using Dunn’s test (AD); Student’s t test (E and F) (*P < 0.05, **P < 0.01, ***P < 0.001).
Figure 4. Ketogenic diet shifts the microbiota…
Figure 4. Ketogenic diet shifts the microbiota composition.
(A) PCoA representing the differences in β diversity of fecal microbiota between dietary interventions normal diet (ND) versus Ketogenic diet (KD) at day 12 in RET–tumor bearing mice. ANOSIM and PERMANOVA defines the separation of the groups; the P value defines the significance of such separation after 999 permutations of the samples. (B) For each principal coordinate axis (PCo1 and PCo2), the collected variance, the Pearson r coefficient and the corresponding P value are shown. Partial Least Squares Discriminant Analysis (PLS-DA) plot of the variance between KD- and ND-fed tumor-bearing mice. LEfSe plot of species at day 12 discriminating ND- from KD-fed tumor-bearing mice, ordered by their LDA score. (C) Detailed relative abundance of distinct species. Refer to Supplemental Figure 4 for additional taxa. (D) Effects of broad-spectrum antibiotics on the percentages of tumor-free mice after ND or KD or 3-hydroxybutyrate per os (3HBpo) after RET inoculation. Data from 2–3 experiments involving 5–6 mice/group are depicted (n = 12 for ND group and n = 21 for KD group). Student t test or ANOVA. **P < 0.01.***P < 0.001.
Figure 5. Correlations between microbial species and…
Figure 5. Correlations between microbial species and ketone bodies in mice and humans.
(AC) Mouse data. Alignment of the taxa relevant in our mouse preclinical studies and the plasma ketone bodies (KB) and Spearman correlations between 2 parameters. The significant ones are indicated with an asterisk (P < 0.05), and their raw data are presented in B and C (each dot represents 1 mouse). (D) Human data. Heatmap showing a correlation matrix between estimates of carbohydrate intake from food questionnaire (FQ) and KB monitoring in plasma in > 1000 individuals from the PREDICT-1 study. The taxa highly significant in our mouse models are highlighted. Pearson correlation was used.
Figure 6. T cell–dependent effects of ketogenic…
Figure 6. T cell–dependent effects of ketogenic diet and synergy with immune checkpoint blockade.
(A) Effects of T cell depletion on tumor sizes at day 12 of diet interventions. Tumor sizes in all diet-fed (normal diet [ND], ketogenic diet [KD], or 3-hydroxybutyrate per os [3HBpo]) groups in the presence of i.p. injections of anti-CD4 and anti-CD8 depleting Abs prior to tumor inoculation. A representative experiment out of 2 is depicted, both yielding similar conclusions; each dot represents 1 mouse, and each group contained 5–6 mice. Mann Whitney U statistical analyses was used. *P < 0.05. (B) Experimental setting for combinatorial regimen. (C and D) Effects of diet interventions on tumor size (C) at day 9 after RET inoculation and diet intervention, after only 1 i.p. administration of cICB (anti–CTLA-4 plus anti–PD-1 mAbs) and overall survival (D). Results of 4 concatenated experiments are depicted; each dot represents 1 mouse (n = 30 for ND and KD groups; n = 12 for 3HBpo and 3HBip). (C) Fold increase of tumor sizes in cICB groups fed with ND, KD, or 3HBpo or 3-hydroxybutyrate i.p. (3HBip) (normalized to ND groups). (D) Overall survival appreciated with Kaplan Meier curves for 12 animals/group after a complete treatment combining continuous diet plus cICB, according to B. (E) Flow cytometry analyses of Tc1 (defined as CXCR3+CD8+ T splenocytes) at day 9 of the combinatorial regimen; each dot represents 1 spleen. Results from 2 pooled experiments are shown. (F) Effects of sucrose supplementation on the synergy between diets and cICB against RET tumor progression. Individual tumor sizes at day 19 of 3 concatenated experiments involving 6 mice/group are shown for each diet intervention; each dot represents 1 tumor/mouse (n = 27 for groups without sucrose, n = 28 for groups on sucrose). Student’s t test (A); global comparison using Kruskall-Wallis test and post-hoc multiple comparisons using Dunn’s test (C and EF). *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 7. Intermittent 3HB scheduling affects systemic…
Figure 7. Intermittent 3HB scheduling affects systemic expression of T cell inhibitory receptor and their ligands.
(A and B) Experimental setting for intemittent diet and combination immune checkpoint blockers (cICB) in the RET model and Kaplan Meier curves of overall survival for 12 animals/group, according to therapeutic scheme comparing continuous (Cont) versus intermittent (On/off) diet interventions. (C) Flow cytometric analyses of PD-1 and CTLA-4 expression in splenic CD8+ T lymphocytes at day 17, after diet (ketogenic diet [KD] or 3-hydroxybutyrate [3HB] Cont or On/off) and systemic cICB combinatorial regimen in RENCA bearing BALB/c mice. Refer to Supplemental Figure 5, A and B, for Tim-3, Lag-3, and 4-1BB in all splenic T cells. (D and E) Flow cytometry analysis of MFI for the membrane expression of costimulatory (CD86 in D) or inhibitory (PD-L1 in E) on macrophages (D) and other myeloid subsets from the spleens at day 17, after complete diet (3HB On/off) and systemic cICB combinatorial regimen in RENCA-bearing BALB/c mice. The results from 2–3 experiments involving 6 mice/group are depicted; each dot represents 1 spleen. (F and G) In vitro effects of increasing dosing of 3HB onto a DC cell line stimulated or not with rIFN-γ. Flow cytometry determination of MHC II/I-Ab (F) and PD-L1 expression (G) as mean fluorescence intensity (MFI) on the surface expression at 48 hours of stimulation. One representative experiment out of 4 is depicted, yielding similar conclusions (n = 12 or each groups). Student’s t test (C), global comparison using Kruskall-Wallis test with post hoc multiple comparisons using Dunn’s test (*P < 0.05) (D and E). Global comparison using ANOVA with post hoc multiple comparisons using Dunnett’s test (*P < 0.05) (F and G). *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 8. Efficacy of intermittent 3HB in…
Figure 8. Efficacy of intermittent 3HB in circumventing primary resistance to PD-1 blockade in an orthotopic cancer model.
(AF) Extension of the comparison between continuous (Cont) versus intermittent (On/off) feeding with KD or 3HBpo in 2 tumor models (RENCA [A], TC-1 [BD]) treated with various therapeutic mAbs (refer to Supplemental Figure 6, A and B, for experimental setting designs). Cross-sectional RENCA tumor burden monitored by bioluminescence imaging of luciferase activity (ratio of luminescence at D8 versus D0 [D8: day of randomization]) (A) are depicted, gathering 2 independent experiments containing 6 mice/group. All experiments were performed twice. Monitoring of TC-1 progression using bioluminescence imaging (B) and percentages of TC-1_luc tumor–free animals overtime monitored by bioluminescence imaging of luciferase activity (C) and overall survival (D) estimated by Kaplan Meier curves for 2 experiments of 6 mice/group pooled together. (E and F) Tumor growth curves represented as mean ± SEM of tumor sizes for each group after s.c. rechallenge of TC-1_luc tumor–free mice (from BD) with the MTD of TC-1_luc (E) or irrelevant MCA205 (F) (refer to Supplemental Figure 6C for experimental setting designs). Experiments were performed twice (n = 12 per groups). Global comparison for the log10-normalized data using ANOVA with post hoc multiple comparisons using Sidak’s test (*P < 0.05) (A) or dedicated software (https://kroemerlab.shinyapps.io/TumGrowth/) (CF). *P < 0.05, **P < 0.01, ***P < 0.001.

References

    1. Wei SC, et al. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069–1086. doi: 10.1158/-18-0367.
    1. Topalian S, et al. Immunotherapy: the path to win the war on cancer? Cell. 2015;161(2):185–186. doi: 10.1016/j.cell.2015.03.045.
    1. Marin-Acevedo JA, et al. Immune checkpoint inhibitor toxicities. Mayo Clin Proc. 2019;94(7):1321–1329. doi: 10.1016/j.mayocp.2019.03.012.
    1. Corrales L, et al. Innate immune signaling and regulation in cancer immunotherapy. Cell Res. 2017;27(1):96–108. doi: 10.1038/cr.2016.149.
    1. Spranger S, Gajewski TF. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer. 2018;18(3):139–147. doi: 10.1038/nrc.2017.117.
    1. Spranger S, Gajewski TF. Tumor-intrinsic oncogene pathways mediating immune avoidance. Oncoimmunology. 2016;5(3):1086862.
    1. Gajewski TF. The next hurdle in cancer immunotherapy: overcoming the non-T-cell-inflamed tumor microenvironment. Semin Oncol. 2015;42(4):663–671. doi: 10.1053/j.seminoncol.2015.05.011.
    1. Zitvogel L, et al. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science. 2018;359(6382):1366–1370. doi: 10.1126/science.aar6918.
    1. Di Sebastiano KM, et al. Nutrition and cancer prevention: why is the evidence lost in translation? Adv Nutr. 2019;10(3):410–418. doi: 10.1093/advances/nmy089.
    1. Sofi F, et al. Adherence to Mediterranean diet and health status: meta-analysis. BMJ. 2008;337:a1344.
    1. Madeo F, et al. Caloric restriction mimetics: towards a molecular definition. Nat Rev Drug Discov. 2014;13(10):727–740. doi: 10.1038/nrd4391.
    1. Pietrocola F, et al. Caloric restriction mimetics enhance anticancer immunosurveillance. Cancer Cell. 2016;30(1):147–160. doi: 10.1016/j.ccell.2016.05.016.
    1. Rubio-Patiño C, et al. Low-protein diet induces IRE1α-dependent anticancer immunosurveillance. Cell Metab. 2018;27(4):828–842. doi: 10.1016/j.cmet.2018.02.009.
    1. Peng W, et al. Surgical stress resistance induced by single amino acid deprivation requires Gcn2 in mice. Sci Transl Med. 2012;4(118):118ra11.
    1. Branco AF, et al. Ketogenic diets: from cancer to mitochondrial diseases and beyond. Eur J Clin Invest. 2016;46(3):285–298. doi: 10.1111/eci.12591.
    1. Klement RJ, Kämmerer U. Is there a role for carbohydrate restriction in the treatment and prevention of cancer? Nutr Metab (Lond) 2011;8(1):75. doi: 10.1186/1743-7075-8-75.
    1. Derosa L, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol. 2020;78(2):195–206. doi: 10.1016/j.eururo.2020.04.044.
    1. Kraeuter A-K, et al. Protocol for the use of the ketogenic diet in preclinical and clinical practice. Methods Mol Biol. 2020;2138:83–98. doi: 10.1007/978-1-0716-0471-7_4.
    1. Warburg O, et al. The metabolism of tumors in the body. J Gen Physiol. 1927;8(6):519–530. doi: 10.1085/jgp.8.6.519.
    1. Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13(6):472–482. doi: 10.1016/j.ccr.2008.05.005.
    1. Baserga R. The contradictions of the insulin-like growth factor 1 receptor. Oncogene. 2000;19(49):5574–5581. doi: 10.1038/sj.onc.1203854.
    1. DeBerardinis RJ, et al. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7(1):11–20. doi: 10.1016/j.cmet.2007.10.002.
    1. Robey RB, Hay N. Is Akt the “Warburg kinase”?—Akt-energy metabolism interactions and oncogenesis. Semin Cancer Biol. 2009;19(1):25–31. doi: 10.1016/j.semcancer.2008.11.010.
    1. Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9(8):550–562. doi: 10.1038/nrc2664.
    1. Zhou W, et al. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond) 2007;4(1):5. doi: 10.1186/1743-7075-4-5.
    1. Meidenbauer JJ, et al. The glucose ketone index calculator: a simple tool to monitor therapeutic efficacy for metabolic management of brain cancer. Nutr Metab (Lond) 2015;12(1):12. doi: 10.1186/s12986-015-0009-2.
    1. Tisdale MJ, Brennan RA. Loss of acetoacetate coenzyme A transferase activity in tumours of peripheral tissues. Br J Cancer. 1983;47(2):293–297. doi: 10.1038/bjc.1983.38.
    1. Skinner R, et al. Ketone bodies inhibit the viability of human neuroblastoma cells. J Pediatr Surg. 2009;44(1):212–216. doi: 10.1016/j.jpedsurg.2008.10.042.
    1. Sourbron J, et al. Ketogenic diet for the treatment of pediatric epilepsy: review and meta-analysis. Childs Nerv Syst. 2020;36(6):1099–1109. doi: 10.1007/s00381-020-04578-7.
    1. Armeno M, Caraballo R. The evolving indications of KD therapy. Epilepsy Res. 2020;163:106340.
    1. Fomin DA, Handfield K. The ketogenic diet and dermatology: a primer on current literature. Cutis. 2020;105(1):40–43.
    1. Caplliure-Llopis J, et al. Therapeutic alternative of the ketogenic Mediterranean diet to improve mitochondrial activity in Amyotrophic Lateral Sclerosis (ALS): a comprehensive review. Food Sci Nutr. 2020;8(1):23–35. doi: 10.1002/fsn3.1324.
    1. Kroemer G, et al. Carbotoxicity-noxious effects of carbohydrates. Cell. 2018;175(3):605–614. doi: 10.1016/j.cell.2018.07.044.
    1. Gopalakrishnan V, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi: 10.1126/science.aan4236.
    1. Lévesque S, et al. A synergistic triad of chemotherapy, immune checkpoint inhibitors, and caloric restriction mimetics eradicates tumors in mice. Oncoimmunology. 2019;8(11):1657375.
    1. Caminhotto R de O, et al. Oral β-hydroxybutyrate increases ketonemia, decreases visceral adipocyte volume and improves serum lipid profile in Wistar rats. Nutr Metab (Lond) 2017;14:31.
    1. Cahill GF. Starvation in man. N Engl J Med. 1970;282(12):668–675.
    1. Laffel L. Ketone bodies: a review of physiology, pathophysiology and application of monitoring to diabetes. Diabetes Metab Res Rev. 1999;15(6):412–426. doi: 10.1002/(SICI)1520-7560(199911/12)15:6<412::AID-DMRR72>;2-8.
    1. Mitchell GA, et al. Medical aspects of ketone body metabolism. Clin Invest Med. 1995;18(3):193–216.
    1. Kim CH. Immune regulation by microbiome metabolites. Immunology. 2018;154(2):220–229. doi: 10.1111/imm.12930.
    1. Berry S, et al. Predicting personal metabolic responses to food using multi-omics machine learning in over 1000 twins and singletons from the UK and US: The PREDICT I Study (OR31-01-19) Curr Dev Nutr. 2019;3(Suppl 1):nzz037
    1. Routy B, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi: 10.1126/science.aan3706.
    1. Groom JR, Luster AD. CXCR3 in T cell function. Exp Cell Res. 2011;317(5):620–631. doi: 10.1016/j.yexcr.2010.12.017.
    1. Curiel TJ, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9(5):562–567. doi: 10.1038/nm863.
    1. Lin H, Wei S, Hurt EM, et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade–mediated tumor regression. J Clin Invest. 2018;128(2):805–815. doi: 10.1172/JCI96113.
    1. Tang H, et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest. 2018;128(2):580–588. doi: 10.1172/JCI96061.
    1. Goldberg EL, et al. Ketogenic diet activates protective γδ T cell responses against influenza virus infection. Sci Immunol. 2019;4(41):eaav2026. doi: 10.1126/sciimmunol.aav2026.
    1. Olson CA, et al. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell. 2018;173(7):1728–1741. doi: 10.1016/j.cell.2018.04.027.
    1. Fluckiger A, et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science. 2020;369(6506):936–942. doi: 10.1126/science.aax0701.
    1. Stekovic S, et al. Alternate day fasting improves physiological and molecular markers of aging in healthy, non-obese humans. Cell Metab. 2019;30(3):462–476. doi: 10.1016/j.cmet.2019.07.016.
    1. Heilbronn LK, et al. Alternate-day fasting in nonobese subjects: effects on body weight, body composition, and energy metabolism. Am J Clin Nutr. 2005;81(1):69–73. doi: 10.1093/ajcn/81.1.69.
    1. Trepanowski JF, et al. Effect of alternate-day fasting on weight loss, weight maintenance, and cardioprotection among metabolically healthy obese adults: a randomized clinical trial. JAMA Intern Med. 2017;177(7):930–938. doi: 10.1001/jamainternmed.2017.0936.
    1. Newman JC, et al. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell Metab. 2017;26(3):547–557. doi: 10.1016/j.cmet.2017.08.004.
    1. McSwiney FT, et al. Keto-adaptation enhances exercise performance and body composition responses to training in endurance athletes. Metabolism. 2018;83:e1–e2.
    1. Woolf EC, et al. Tumor metabolism, the ketogenic diet and β-hydroxybutyrate: novel approaches to adjuvant brain tumor therapy. Front Mol Neurosci. 2016;9:122.
    1. Cohen CW, et al. A ketogenic diet reduces central obesity and serum insulin in women with ovarian or endometrial cancer. J Nutr. 2018;148(8):1253–1260. doi: 10.1093/jn/nxy119.
    1. Snell-Bergeon JK, et al. Adults with type 1 diabetes eat a high-fat atherogenic diet that is associated with coronary artery calcium. Diabetologia. 2009;52(5):801–809. doi: 10.1007/s00125-009-1280-4.
    1. Kim D, et al. Monitoring the trafficking of adoptively transferred antigen- specific CD8-positive T cells in vivo, using noninvasive luminescence imaging. Hum Gene Ther. 2007;18(7):575–588. doi: 10.1089/hum.2007.038.
    1. Zhou W, et al. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond) 2007;4:5.
    1. Dridi B, et al. High prevalence of methanobrevibacter smithii and methanosphaera stadtmanae detected in the human gut using an improved DNA detection protocol. PLoS One. 2009;4(9):e7063. doi: 10.1371/journal.pone.0007063.
    1. Angelakis E, et al. Glycans affect DNA extraction and induce substantial differences in gut metagenomic studies. Sci Rep. 2016;6:26276.
    1. Million M, et al. Increased gut redox and depletion of anaerobic and methanogenic prokaryotes in severe acute malnutrition. Sci Rep. 2016;6:26051.
    1. Salem HA, Wadie W. Effect of niacin on inflammation and angiogenesis in a murine model of ulcerative colitis. Sci Rep. 2017;7(1):7139. doi: 10.1038/s41598-017-07280-y.
    1. Richter C, et al. Generation of inducible immortalized dendritic cells with proper immune function in vitro and in vivo. PLoS One. 2013;8(4):62621.
    1. R. The R Project for Statistical Computing. Accessed December 15, 2020.
    1. GraphPad. QuickCalcs: outlier calculator. Accessed December 15, 2020.
    1. Enot DP, et al. TumGrowth: An open-access web tool for the statistical analysis of tumor growth curves. Oncoimmunology. 2018;7(9):1462431.
    1. TumGrowth. Analysis of tumour growth experiments. Accessed December 15, 2020.

Source: PubMed

3
Tilaa