Fasting-Mimicking Diet Is Safe and Reshapes Metabolism and Antitumor Immunity in Patients with Cancer

Claudio Vernieri, Giovanni Fucà, Francesca Ligorio, Veronica Huber, Andrea Vingiani, Fabio Iannelli, Alessandra Raimondi, Darawan Rinchai, Gianmaria Frigè, Antonino Belfiore, Luca Lalli, Claudia Chiodoni, Valeria Cancila, Federica Zanardi, Arta Ajazi, Salvatore Cortellino, Viviana Vallacchi, Paola Squarcina, Agata Cova, Samantha Pesce, Paola Frati, Raghvendra Mall, Paola Antonia Corsetto, Angela Maria Rizzo, Cristina Ferraris, Secondo Folli, Marina Chiara Garassino, Giuseppe Capri, Giulia Bianchi, Mario Paolo Colombo, Saverio Minucci, Marco Foiani, Valter Daniel Longo, Giovanni Apolone, Valter Torri, Giancarlo Pruneri, Davide Bedognetti, Licia Rivoltini, Filippo de Braud, Claudio Vernieri, Giovanni Fucà, Francesca Ligorio, Veronica Huber, Andrea Vingiani, Fabio Iannelli, Alessandra Raimondi, Darawan Rinchai, Gianmaria Frigè, Antonino Belfiore, Luca Lalli, Claudia Chiodoni, Valeria Cancila, Federica Zanardi, Arta Ajazi, Salvatore Cortellino, Viviana Vallacchi, Paola Squarcina, Agata Cova, Samantha Pesce, Paola Frati, Raghvendra Mall, Paola Antonia Corsetto, Angela Maria Rizzo, Cristina Ferraris, Secondo Folli, Marina Chiara Garassino, Giuseppe Capri, Giulia Bianchi, Mario Paolo Colombo, Saverio Minucci, Marco Foiani, Valter Daniel Longo, Giovanni Apolone, Valter Torri, Giancarlo Pruneri, Davide Bedognetti, Licia Rivoltini, Filippo de Braud

Abstract

In tumor-bearing mice, cyclic fasting or fasting-mimicking diets (FMD) enhance the activity of antineoplastic treatments by modulating systemic metabolism and boosting antitumor immunity. Here we conducted a clinical trial to investigate the safety and biological effects of cyclic, five-day FMD in combination with standard antitumor therapies. In 101 patients, the FMD was safe, feasible, and resulted in a consistent decrease of blood glucose and growth factor concentration, thus recapitulating metabolic changes that mediate fasting/FMD anticancer effects in preclinical experiments. Integrated transcriptomic and deep-phenotyping analyses revealed that FMD profoundly reshapes anticancer immunity by inducing the contraction of peripheral blood immunosuppressive myeloid and regulatory T-cell compartments, paralleled by enhanced intratumor Th1/cytotoxic responses and an enrichment of IFNγ and other immune signatures associated with better clinical outcomes in patients with cancer. Our findings lay the foundations for phase II/III clinical trials aimed at investigating FMD antitumor efficacy in combination with standard antineoplastic treatments. SIGNIFICANCE: Cyclic FMD is well tolerated and causes remarkable systemic metabolic changes in patients with different tumor types and treated with concomitant antitumor therapies. In addition, the FMD reshapes systemic and intratumor immunity, finally activating several antitumor immune programs. Phase II/III clinical trials are needed to investigate FMD antitumor activity/efficacy.This article is highlighted in the In This Issue feature, p. 1.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
The fasting-mimicking diet (FMD) reduces blood glucose and growth factor levels in patients with cancer. A, Schematics of the FMD regimen, with the calorie content of each day of FMD (d1–d5). Black arrows indicate time points of blood/urine sample collection. Blood samples collected before the initiation of the FMD are indicated as “Pre,” whereas samples collected after the end of five-day FMD (i.e., before the initiation of refeeding) are indicated as “Post.” B, Bar plots indicating the number of patients (y-axis) who completed the number of FMD cycles indicated on the x-axis. C, Concentration of plasma glucose (mg/dL), serum insulin (10*μUI/mL), serum IGF1 (ng/mL), and urinary ketone bodies (mg/dL) before (Pre) and after (Post) FMD (first cycle) in 99 patients with cancer. D, Plasma glucose concentration (mg/dL) before and after FMD (across eight cycles; C1–C8). E, Serum insulin concentration (μUI/mL) before and after FMD (across eight cycles; C1–C8). F, Serum IGF1 concentration (ng/mL) before and after FMD (across eight cycles; C1–C8). G, BMI before and after FMD cycles (C1–C8) in the whole patient population. H, BMI before and after FMD (C1–C8) in patients undergoing FMD and cytotoxic chemotherapy (ChT). I, BMI before and after FMD (C1–C8) in patients undergoing FMD in combination with treatment other than ChT. All P values were determined by paired Wilcoxon test: *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. All comparisons for which the P value is not indicated did not show statistically significant differences (P ≥ 0.05). Data are represented as box plots showing median values, with the boundaries of the rectangle representing the first and third quartiles, while whiskers extend to the extreme data points that are no more than 1.5 times the interquartile range.
Figure 2.
Figure 2.
The fasting-mimicking diet (FMD) reduces peripheral blood immunosuppressive cells and increases effector cells in patients with cancer and in healthy volunteers. A, Frequencies of CD14+, CD14+HLA-DR−, CD14+PD-L1+, and CD15+ cells before (Pre) and after (Post) FMD (first cycle) in 38 patients with different tumor types enrolled in the NCT03340935 trial. CD14+ and CD15+ cells were calculated as frequencies in total PBMCs (after debris and doublet exclusion), while CD14+PD-L1+ and CD14+HLA-DR− cells were calculated as frequencies in total CD14+ cells. B, Frequencies of CD14+, CD14+HLA-DR−, CD14+PD-L1+, and CD15+ cells before (Pre) and after (Post) FMD (first cycle) in 13 patients with advanced breast cancer enrolled in the NCT03340935 trial and treated with first-line ChT plus FMD. C, Frequencies of CD14+, CD14+HLA-DR−, CD14+PD-L1+ and CD15+ cells before (Pre) and after (Post) ChT in a cohort of 13 patients with advanced breast cancer treated with ChT without the FMD. D, Frequencies of CD14+, CD14+HLA-DR−, CD14+PD-L1+, and CD15+ cells before (Pre) and after (Post) FMD in 8 healthy volunteers. E, Frequencies of pre- and post-FMD CD8+PD1+CD69+ (among CD3+CD69+), CD3+CD25+ (among CD3+), and CD3−CD16+CD56dim (among CD3−) cells in 38 patients with different tumor types enrolled in the NCT03340935 trial. F, Frequencies of pre- and post-FMD CD8+PD1+CD69+ (among CD3+CD69+), CD3+CD25+ (among CD3+) and CD3−CD16+CD56dim (among CD3−) cells in 13 patients with advanced breast cancer enrolled in the NCT03340935 trial and treated with first-line ChT plus FMD. G, Frequencies of CD8+PD-1+CD69+ (among CD3+CD69+), CD3+CD25+ (among CD3+), and CD3−CD16+CD56dim (among CD3−) cells before and after ChT in 13 patients with advanced breast cancer treated with ChT without the FMD. H, Frequencies of pre- and post-FMD CD8+PD-1+CD69+ (among CD3+CD69+), CD3+CD25+ (among CD3+), and CD3−CD16+CD56dim (among CD3−) cells in 8 healthy volunteers. I, Plasma concentration of CCL2, G-CSF, IL6, and IL8, as evaluated at the initiation and at the end of the FMD, in 34 patients with different tumor types enrolled in the NCT03340935 trial. CCL2, C-C motif chemokine ligand 2. All P values were determined by paired Wilcoxon test: *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. All comparisons for which the P value is not indicated did not show statistically significant differences (P ≥ 0.05). Box plots indicate median values of the indicated variable, with the boundaries of the rectangle representing the first and third quartiles, while vertical black lines extend to the extreme data points that are no more than 1.5 times the interquartile range. Each red dot represents one patient; couples of dots connected by the same black line refer to data from the same patient before and after the FMD.
Figure 3.
Figure 3.
The fasting-mimicking diet (FMD) reshapes intratumor immunity in patients with breast cancer. A, Time points of blood and tissue sample collection in the DigesT trial (NCT03454282). T1, initiation of the FMD; T2, end of the FMD; T3, surgery; T4, approximately 30 days after surgery. B, Left, IHC evaluation of intratumor IGFR1 and phosphorylated IGFR1 (pIGF1R; brown dots) in pre- and post-FMD tumor samples from one indicative patient (magnification of 200×). B, Right, Box plots showing results of IHC analyses (reported as H-score) of IGF1R and pIGF1R in matched pre- and post-FMD tumor samples in 18 patients enrolled in the DigesT trial (NCT03454282). All P values for the indicated comparisons were determined by paired Wilcoxon test: *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. Box plots indicate median values, with the boundaries of the rectangle representing the first and third quartiles, while vertical black lines extend to the extreme data points that are no more than 1.5 times the interquartile range. Each red dot represents one patient; couples of plots connected by the same black line refer to the same patient before and after the FMD. C, Representative images of hematoxylin and eosin staining (H&E) and IHC evaluations of intratumor CD8+ T cells and CD68+ macrophages in pre-FMD (T1) and post-FMD (T3) tumor samples from two representative patients (magnification of ×200). D, Top plots, box plots showing the IHC quantification of percentage (area) and absolute average numbers (per high-power field; HPF) of intratumor stromal CD8+ cells, absolute average number (per HPF) of intraepithelial CD8+ cells, absolute average number (per HPF) of CD68+ macrophages, and the ratio between stromal CD8+ cells (absolute average number per HPF) and CD68+ macrophages (absolute average number per HPF) in pre-FMD versus post-FMD tumor specimens. Box plot showing the CD8A/CD68 transcript ratio in post-FMD as compared with pre-FMD tumor specimens. Box plots indicate median values of the frequencies of each immune cell population/gene transcript, with the boundaries of the rectangle representing the first and third quartiles, while vertical black lines extend to the extreme data points that are no more than 1.5 times the interquartile range. Each red dot represents one patient; couples of dots connected by the same black line refer to data from the same patient pre- and post-FMD. *, P < 0.05; **, P < 0.01; ***, P < 0.001 by paired Wilcoxon test for the indicated comparisons. D, Bottom plots, correlation between CD8+ T cells by IHC and CD8A gene expression, and between CD68+ macrophages by IHC and CD68 gene expression; box plot showing the CD8A/CD68 transcript ratio in post-FMD as compared with pre-FMD tumor specimens. E, Heat map displaying scores of enrichment of leukocyte subsets (XCell, Charoentong list) that are differentially modulated after the FMD [T3 vs. T1, paired Wilcoxon test, P < 0.05, Benjamini–Hochberg (B-H) false discovery rate (FDR) < 0.1]. aDC, activated DCs; CD4+ and CD8+ Tcm: CD4+ and CD8+ central memory T lymphocytes; NKT, natural killer T cells; pDC, plasmacytoid dendritic cells; Tem, effector memory T lymphocytes. F, Box plots representing scores of individual leukocyte subsets modulated in post-FMD versus pre-FMD tumor specimens. G, Matrix correlation displaying Spearman correlation coefficients between RNA-seq data (CD8A and CD68 transcripts, CD8+ T-cell, and Macrophage enrichment scores), and IHC assessment of CD8+ T cells and CD68+ cells.
Figure 4.
Figure 4.
Intratumor transcriptomic changes induced by fasting-mimicking diet (FMD). A, Volcano plots displaying differentially expressed genes comparing post-FMD (T3) versus pre-FMD (T1) gene-level RNA-seq data using negative binomial distribution (Deseq2 package). Benjamini–Hochberg (B-H) false discovery rate (FDR) and Log2 fold change are represented. Left, red and blue colors are used to display upregulated and downregulated transcripts in post-FMD versus pre-FMD tumors, respectively; immune-related genes (see Methods and Supplementary Table S10) are colored in dark red (upregulated in post-FMD versus pre-FMD comparison) and dark blue (downregulated in post-FMD versus pre-FMD comparison); all the immune-related genes with −log10 FDR > 2.5 are labeled; additional representative immune-related genes with −log10 FDR > 1 are labeled; in the middle and left panels, transcripts belonging to the IFN-activating signature (IFNG.GS) and IFN-resistant signature (ISG.RS) are colored in purple and green, respectively. Volcano plot y-axis is truncated at −log10 FDR = 10, as no immune-related genes above that cutoff were present. B, Heat maps representing differentially expressed genes in post-FMD (T3, surgical samples) versus pre-FMD (T1, tumor biopsies) comparisons; the top panel displays genes with P < 0.05 (Wilcoxon test) and B-H FDR < 0.1 at differential gene expression analysis and included in any of the significantly modulated (P < 0.05 and B-H FDR < 0.1) prognostic/predictive signatures at single-sample gene set enrichment analysis (of the 25 evaluated prognostic/predictive signatures, 13 were significantly enriched in post-FMD vs. pre-FMD tumor samples, but one of them did not yield any differentially expressed genes, therefore only 12 signatures are represented in the heat map; Supplementary Table S10); the middle panel displays genes with P < 0.05 (Wilcoxon test) and B-H FDR < 0.1 at differential gene expression analysis and included in the exhaustion and immune checkpoint list (Supplementary Table S10); the bottom panel displays genes with P < 0.05 and B-H FDR < 0.1 at differential gene expression analysis and included in the cytokine/chemokine gene list (Supplementary Table S10). In the heat maps, all genes with B-H FDR < 0.05 are labeled, in addition with other selected genes (in brackets) with FDR between 0.1 and 0.05. C, Box plots of representative prognostic/predictive signature enrichment scores and transcripts with P < 0.05 and B-H FDR < 0.1 in the post-FMD (T3) versus pre-FMD (T1) comparison. D, Box plots of Perforin 1 and Granzyme B IHC scores in post-FMD (T3) versus pre-FMD (T1) tumors (n = 17 and n = 16, respectively).
Figure 5.
Figure 5.
Dynamic effects of FMD on systemic immunity and correlation with transcriptional immune modulation at tumor site. High-dimensional flow cytometry was performed in PBMCs from 19 of 22 patients enrolled in the DigesT trial selected for tumor RNA-seq analysis. A, Samples for high-dimensional flow cytometry were collected before the FMD (T1) and at different time points after the FMD (T2, T3, T4). We assessed the expression of multiple lymphoid and myeloid markers to define a total of 120 cell subsets (see Supplementary Fig. S11–S13). The nonparametric paired Wilcoxon test was used to compare the diverse immune cell frequencies at T2 versus T1, T3 versus T1, and T4 versus T1; 57 PBMC subpopulations undergoing modifications passing the significance cutoff at least at one time point (P < 0.05, Benjamini-Hochberg FDR < 0.1) are represented in the heat map. B, Loess regression curves of representative immune cell subsets are shown to illustrate modulation across time. See Supplementary Fig. S14 for the representation of the remaining PBMC Loess regression curves related to the significantly modulated PBMC subsets. C, Heat map representing Spearman correlations between significantly modulated blood PBMC subpopulations (n = 57) and intratumor leucocyte subsets (n = 14) at T3 versus T1 in 19 patients for whom both PBMC flow cytometry and tumor RNA-seq data were available. D, Representative correlations between individual patient values of the indicated PBMC and intratumor leukocyte subsets (P < 0.05, Benjamini–Hochberg FDR < 0.1) at T1 and T3.

References

    1. Lee C, Raffaghello L, Brandhorst S, Safdie FM, Bianchi G, Martin-Montalvo A, et al. . Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med 2012;4:124ra27.
    1. Lee C, Safdie FM, Raffaghello L, Wei M, Madia F, Parrella E, 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 2010;70:1564–72.
    1. Di Biase S, Lee C, Brandhorst S, Manes B, Buono R, Cheng CW, et al. . Fasting-mimicking diet reduces HO-1 to promote T cell-mediated tumor cytotoxicity. Cancer Cell 2016;30:136–46.
    1. Di Tano M, Raucci F, Vernieri C, Caffa I, Buono R, Fanti M, et al. . Synergistic effect of fasting-mimicking diet and vitamin C against KRAS mutated cancers. Nat Commun 2020;11:2332.
    1. Ajona D, S. O-E, Lozano T, Exposito F, Calvo A, Valencia K, et al. . Short-term starvation reduces IGF-1 levels to sensitize lung tumors to PD-1 immune checkpoint blockade. Nat Cancer 2020;1:75–85.
    1. Caffa I, Spagnolo V, Vernieri C, Valdemarin F, Becherini P, Wei M, et al. . Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 2020;583:620–4.
    1. de Groot S, Lugtenberg RT, Cohen D, Welters MJP, Ehsan I, Vreeswijk MPG, 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;11:3083.
    1. Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, et al. . Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 2016;7:12150.
    1. Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M, et al. . Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 2007;25:2546–53.
    1. Roux C, Jafari SM, Shinde R, Duncan G, Cescon DW, Silvester J, et al. . Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1. Proc Natl Acad Sci U S A 2019;116:4326–35.
    1. Moretta L. Dissecting CD56dim human NK cells. Blood 2010;116:3689–91.
    1. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016;44:989–1004.
    1. DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, et al. . Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov 2011;1:54–67.
    1. Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. . Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep 2017;18:248–62.
    1. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol 2017;18:220.
    1. Benci JL, Johnson LR, Choa R, Xu Y, Qiu J, Zhou Z, et al. . Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell 2019;178:933–48.
    1. Condamine T, Dominguez GA, Youn JI, Kossenkov AV, Mony S, Alicea-Torres K, et al. . Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol 2016;1:aaf8943.
    1. Krieg C, Nowicka M, Guglietta S, Schindler S, Hartmann FJ, Weber LM, et al. . High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med 2018;24:144–53.
    1. Wang L, Simons DL, Lu X, Tu TY, Avalos C, Chang AY, et al. . Breast cancer induces systemic immune changes on cytokine signaling in peripheral blood monocytes and lymphocytes. EBioMedicine 2020;52:102631.
    1. Fromm PD, Silveira PA, Hsu JL, Papadimitrious MS, Lo TH, Ju X, et al. . Distinguishing human peripheral blood CD16(+) myeloid cells based on phenotypic characteristics. J Leukoc Biol 2020;107:323–39.
    1. Gerlach C, Moseman EA, Loughhead SM, Alvarez D, Zwijnenburg AJ, Waanders L, et al. . The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity 2016;45:1270–84.
    1. Yan Y, Cao S, Liu X, Harrington SM, Bindeman WE, Adjei AA, et al. . CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy. JCI Insight 2018;3:e97828.
    1. Flippe L, Bezie S, Anegon I, Guillonneau C. Future prospects for CD8(+) regulatory T cells in immune tolerance. Immunol Rev 2019;292:209–24.
    1. André F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. . Alpelisib for PIK3CA-mutated, hormone receptor–positive advanced breast cancer. N Engl J Med 2019;380:1929–40.
    1. Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. . Pembrolizumab plus chemotherapy in metastatic non–small-cell lung cancer. N Engl J Med 2018;378:2078–92.
    1. Cremolini C, Antoniotti C, Rossini D, Lonardi S, Loupakis F, Pietrantonio F, et al. . Upfront FOLFOXIRI plus bevacizumab and reintroduction after progression versus mFOLFOX6 plus bevacizumab followed by FOLFIRI plus bevacizumab in the treatment of patients with metastatic colorectal cancer (TRIBE2): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol 2020;21:497–507.
    1. Vernieri C, Ligorio F, Zattarin E, Rivoltini L, de Braud F. Fasting-mimicking diet plus chemotherapy in breast cancer treatment. Nat Commun 2020;11:4274.
    1. Argiles JM, Busquets S, Stemmler B, Lopez-Soriano FJ. Cancer cachexia: understanding the molecular basis. Nat Rev Cancer 2014;14:754–62.
    1. Elgendy M, Ciro M, Hosseini A, Weiszmann J, Mazzarella L, Ferrari E, et al. . Combination of hypoglycemia and metformin impairs tumor metabolic plasticity and growth by modulating the PP2A-GSK3beta-MCL-1 axis. Cancer Cell 2019;35:798–815.
    1. Vernieri C, Casola S, Foiani M, Pietrantonio F, de Braud F, Longo V. Targeting cancer metabolism: dietary and pharmacologic interventions. Cancer Discov 2016;6:1315–33.
    1. Dorff TB, Groshen S, Garcia A, Shah M, Tsao-Wei D, Pham H, et al. . Safety and feasibility of fasting in combination with platinum-based chemotherapy. BMC Cancer 2016;16:360.
    1. Bauersfeld SP, Kessler CS, Wischnewsky M, Jaensch A, Steckhan N, Stange R, et al. . The effects of short-term fasting on quality of life and tolerance to chemotherapy in patients with breast and ovarian cancer: a randomized cross-over pilot study. BMC Cancer 2018;18:476.
    1. de Groot S, Vreeswijk MP, Welters MJ, Gravesteijn G, Boei JJ, Jochems A, et al. . The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy in HER2-negative breast cancer patients: a randomized pilot study. BMC Cancer 2015;15:652.
    1. Ferrere G, Tidjani Alou M, Liu P, Goubet AG, Fidelle M, Kepp O, et al. . Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight 2021;6:e145207.
    1. Jordan S, Tung N, Casanova-Acebes M, Chang C, Cantoni C, Zhang D, et al. . Dietary intake regulates the circulating inflammatory monocyte pool. Cell 2019;178:1102–14.
    1. Li W, Tanikawa T, Kryczek I, Xia H, Li G, Wu K, et al. . Aerobic glycolysis controls myeloid-derived suppressor cells and tumor immunity via a specific CEBPB isoform in triple-negative breast cancer. Cell Metab 2018;28:87–103.
    1. De Rosa V, Galgani M, Porcellini A, Colamatteo A, Santopaolo M, Zuchegna C, et al. . Glycolysis controls the induction of human regulatory T cells by modulating the expression of FOXP3 exon 2 splicing variants. Nat Immunol 2015;16:1174–84.
    1. Bilbao D, Luciani L, Johannesson B, Piszczek A, Rosenthal N. Insulin-like growth factor-1 stimulates regulatory T cells and suppresses autoimmune disease. EMBO Mol Med 2014;6:1423–35.
    1. Newton R, Priyadharshini B, Turka LA. Immunometabolism of regulatory T cells. Nat Immunol 2016;17:618–25.
    1. Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, et al. . LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab 2016;24:657–71.
    1. Ho PC, Bihuniak JD, Macintyre AN, Staron M, Liu X, Amezquita R, et al. . Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 2015;162:1217–28.
    1. Cascone T, McKenzie JA, Mbofung RM, Punt S, Wang Z, Xu C, et al. . Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab 2018;27:977–87.
    1. Collins N, Han SJ, Enamorado M, Link VM, Huang B, Moseman EA, et al. . The bone marrow protects and optimizes immunological memory during dietary restriction. Cell 2019;178:1088–101.
    1. Mlecnik B, Tosolini M, Charoentong P, Kirilovsky A, Bindea G, Berger A, et al. . Biomolecular network reconstruction identifies T-cell homing factors associated with survival in colorectal cancer. Gastroenterology 2010;138:1429–40.
    1. Bedognetti D, Spivey TL, Zhao Y, Uccellini L, Tomei S, Dudley ME, et al. . CXCR3/CCR5 pathways in metastatic melanoma patients treated with adoptive therapy and interleukin-2. Br J Cancer 2013;109:2412–23.
    1. Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham R, et al. . Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 2021;184:596–614.
    1. Galon J, Angell HK, Bedognetti D, Marincola FM. The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures. Immunity 2013;39:11–26.
    1. Orillion A, Damayanti NP, Shen L, Adelaiye-Ogala R, Affronti H, Elbanna M, et al. . Dietary protein restriction reprograms tumor-associated macrophages and enhances immunotherapy. Clin Cancer Res 2018;24:6383–95.
    1. Rubio-Patino C, Bossowski JP, De Donatis GM, Mondragon L, Villa E, Aira LE, et al. . Low-protein diet induces IRE1alpha-dependent anticancer immunosurveillance. Cell Metab 2018;27:828–42.
    1. Green SJ, Dahlberg S. Planned versus attained design in phase II clinical trials. Stat Med 1992;11:853–62.

Source: PubMed

3
Abonner