Preexisting Immunity Drives the Response to Neoadjuvant Chemotherapy in Esophageal Adenocarcinoma

Giuseppina Arbore, Luca Albarello, Gabriele Bucci, Marco Punta, Andrea Cossu, Lorella Fanti, Aurora Maurizio, Francesco Di Mauro, Vito Bilello, Gianluigi Arrigoni, Silvia Bonfiglio, Donatella Biancolini, Francesco Puccetti, Ugo Elmore, Luca Vago, Stefano Cascinu, Giovanni Tonon, Riccardo Rosati, Giulia Casorati, Paolo Dellabona, Giuseppina Arbore, Luca Albarello, Gabriele Bucci, Marco Punta, Andrea Cossu, Lorella Fanti, Aurora Maurizio, Francesco Di Mauro, Vito Bilello, Gianluigi Arrigoni, Silvia Bonfiglio, Donatella Biancolini, Francesco Puccetti, Ugo Elmore, Luca Vago, Stefano Cascinu, Giovanni Tonon, Riccardo Rosati, Giulia Casorati, Paolo Dellabona

Abstract

Current treatment for patients with locally advanced esophageal adenocarcinoma (EAC) is neoadjuvant chemotherapy (nCT), alone or combined with radiotherapy, before surgery. However, fewer than 30% of treated patients show a pathologic complete response to nCT, which correlates with increased 5-year survival compared with nonresponders. Understanding the mechanisms of response to nCT is pivotal to better stratify patients and inform more efficacious therapies. Here, we investigated the immune mechanisms involved in nCT response by multidimensional profiling of pretreatment tumor biopsies and blood from 68 patients with EAC (34 prospectively and 34 retrospectively collected), comparing complete responders versus nonresponders to nCT. At the tumor level, complete response to nCT was associated with molecular signatures of immune response and proliferation, increased putative antitumor tissue-resident memory CD39+ CD103+ CD8+ T cells, and reduced immunosuppressive T regulatory cells (Treg) and M2-like macrophages. Systemically, complete responders showed higher frequencies of immunostimulatory CD14+ CD11c+ HLA-DRhigh cells, and reduced programmed cell death ligand 1-positive (PD-L1+) monocytic myeloid-derived suppressor cells, along with high plasma GM-CSF (proinflammatory) and low IL4, CXCL10, C3a, and C5a (suppressive). Plasma proinflammatory and suppressive cytokines correlated directly and inversely, respectively, with the frequency of tumor-infiltrating CD39+ CD103+ CD8+ T cells. These results suggest that preexisting immunity in baseline tumor drives the clinical activity of nCT in locally advanced EAC. Furthermore, it may be possible to stratify patients based on predictive immune signatures, enabling tailored neoadjuvant and/or adjuvant regimens.

Significance: Multidimensional profiling of pretreatment esophageal adenocarcinoma shows patient response to nCT is correlated with active preexisting immunity and indicates molecular pathways of resistance that may be targeted to improve clinical outcomes.

©2023 The Authors; Published by the American Association for Cancer Research.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Baseline higher immune gene signatures and reduced resistance to stress are associated with response to nCT. A, Schematic overview of the study design showing our multidimensional analysis of pre-nCT tumor biopsies (WES, RNA-seq, IHC, proteome DSP, and flow cytometry analysis) and blood (flow cytometry analysis, plasma cytokines) collected prospectively from patients diagnosed with locally advanced EAC (n = 34); an additional cohort of archived pretreatment EAC tumors (n = 34) was analyzed by IHC and DSP. B, Hallmark GSEA comparison of CRs (n = 9) versus NRs (n = 9) showing normalized enrichment scores for gene sets with FDR < 0.01. C, Volcano plot of genes enriched in CRs versus NRs from bulk RNA-seq. The volcano plot reports the name of genes selected on the basis of their participation in pathways differentially enriched in GSEA analysis, as discussed in the Results section. D, Oncoplot of somatic mutations in Reactome pathway MHC class I antigen processing and presentation genes, with respective percentages of mutated genes for each EAC sample. E, HLA LOH events represented as imbalanced HLA copy numbers observed in two patients, N 20 (PR) and N 8 (NR).
Figure 2.
Figure 2.
Increased baseline infiltration of FOXP3+ and CD163+ cells in EACs of NRs is associated with reduced OS. A, CIBERSORT scores for EAC-infiltrating immune cell populations were calculated from RNA-seq data for CRs, PRs, and NRs. Bar graphs represent mean ± SEM. *, P <0.05; **, P <0.01; one-way ANOVA. NV B, naïve B; ME B, memory B; P, plasma cells; CD8, CD8+; NV CD4, naïve CD4+; ME CD4 R, memory CD4+ resting; ME CD4 A, memory CD4+ activated; TFH, follicular T helper; γδT, gamma delta T; NK R, natural killer resting; NK A, natural killer activated; MONO, monocytes; M0, M0 macrophages; M1, M1 macrophages; M2, M2 macrophages; DC R, dendritic cells resting; DC A, dendritic cells activated; MC R, mast cells resting; MC A, mast cells activated; EO, eosinophils; NEU, neutrophils. B, Representative immunostaining of pretreatment EACs for FOXP3, CD163, and CD45RO. Scale bar, 50 μm. C, Staining scores for intratumoral (intra Tum) and peritumoral regions (peri Tum) in CRs (n = 15), PRs (n = 16), and NRs (n = 33). Semiquantitative h scores (0 = no staining to 3 = strongest staining) were assigned by an expert pathologist. Data are presented as mean ± SEM. *, P <0.05; one-way ANOVA. D, OS of patients with high versus low FOXP3 IHC staining scores in intratumor EAC infiltrates before neoadjuvant chemotherapy (high score, h ≥ 1; low score, h < 1). *, P <0.05; log-rank test. E and F, OS of patients with high versus low CD163 IHC staining scores in peritumoral (E) and intratumoral (F) EAC infiltrates before neoadjuvant chemotherapy (high score, h ≥ 2; low score, h < 2). *, P < 0.05; **, P < 0.01; log-rank test.
Figure 3.
Figure 3.
DSP confirms distinct baseline functional immune contexture in EACs of CRs and NRs. A, Experimental design: spatial proteomic profiling of 53 tumor and immune markers was conducted on FFPE EAC biopsy sections; selection of multiple ROIs per tissue sample was based on immunofluorescent staining for PanCK (tumor epithelial marker), CD68 (macrophages), and CD3 (T lymphocytes); nuclei were stained with 4',6-diamidino-2-phenylindole (DAPI). Protein counts were measured within PanCK+-enriched tumor regions (masks) and PanCK− stromal regions (inverted masks). B, Volcano plot of proteins enriched in PanCK+ (n = 72) versus PanCK− (n = 72) areas showing statistical significance for proteins with P < 0.05; P values were calculated using the linear mixed model. C, Representative immunofluorescent staining for PanCK, CD68, and CD3 in tumor tissue from CRs and NRs. Scale bar, 100 μm. D, CD3 PanCK+/PanCK− ratios for each ROI (normalized by area) for CRs and NRs (calculated from DSP reads). *, P <0.05, unpaired t test. E and G, Volcano plots of proteins enriched in PanCK− (E) and PanCK+ (G) areas in tumors from CRs (n = 33 ROIs) versus NRs (n = 39 ROIs), showing statistical significance for proteins with P <0.05; P values were calculated using the linear mixed model. F and H, Corresponding box plots representing read numbers (N) normalized to the area for proteins differentially enriched in PanCK− (F) and PanCK+ areas (H), presented as mean ± SEM. *, P < 0.05; **, P < 0.01, unpaired t test.
Figure 4.
Figure 4.
Responder EACs show reduced baseline infiltration of FOXP3+ Tregs and an increased frequency of tissue-resident CD8+CD39+ T cells. A, Representative flow cytometry plots of tumor single-cell suspensions (CD3+ viable cells) stained for nine surface markers, showing the gating strategy for CD25+ CD127low Tregs and CD8+ CD39+ CD103+ T cells. B and C, Dot plots showing frequencies expressed as percentage of CD25+ CD127low CD4+ cells (B) and CD39+ CD103+ CD8+ cells (C) in pretreatment EAC tumor cell suspensions from CRs (n = 9) and NRs (n = 6), determined by manual gating. Data are presented as mean ± SEM. *, P < 0.05, unpaired t test. D, Exemplified t-SNE visualization of 17 surface markers representing 6,000 merged CD3+ CD4+ cells in tumor cell suspensions from CRs (n = 3) and NRs (n = 3); 13 clusters were predicted by unsupervised PhenoGraph analysis. E, Histograms showing the expression of six surface markers in clusters #4 and #5, concatenated CD3+ CD4+ events (Conc), and negative control (Neg); relative fluorescence intensity (RFI) values are shown. F, Exemplified t-SNE visualization of 17 surface markers representing 6,000 merged CD3+ CD8+ cells in tumor cell suspensions from CRs (n = 3) and NRs (n = 3); 11 clusters were predicted by unsupervised PhenoGraph analysis. G, Histograms showing the expression (RFI values) of 12 surface markers in clusters #1 and #2, concatenated CD3+ CD8+ events, and negative control. H, Exemplified t-SNE visualization of eight markers, including three surface markers and five nuclear transcription factors, representing 6,000 merged CD3+ CD4+ cells in tumor cell suspensions from CRs (n = 3) and NRs (n = 3); nine clusters were predicted by unsupervised PhenoGraph analysis. I, Histograms showing the expression (RFI values) of seven markers in cluster #6, (CD25+ CD127low FOXP3+ Treg-like cells), concatenated CD3+ CD4+ events, and negative control. J, Exemplified t-SNE visualization of 8 markers (three surface markers and five nuclear transcription factors), representing 6,000 merged CD3+ CD4− cells in tumor cell suspensions from CRs (n = 3) and NRs (n = 3); nine clusters were predicted by unsupervised PhenoGraph analysis. K, Histograms showing the expression (RFI values) of eight markers in cluster #3, concatenated CD3+ CD4− events, and negative control.
Figure 5.
Figure 5.
NR EACs show increased baseline infiltration of protumor M2-like macrophages. A, Exemplified tSNE visualization of 20 surface markers representing 7,000 merged CD45+ CD3− tumor cells from CRs (n = 3) and NRs (n = 4); cells are colored according to the 10 clusters assigned by PhenoGraph and manual annotation; the corresponding heatmap shows median marker intensity normalized to a 0 to 1 range. B, Frequencies of the clusters in CR (n = 3) and NR (n = 4) CD45+ CD3− cells presented as mean ± SEM. *, P < 0.05; one-way ANOVA. C, Histograms showing the expression of six surface markers in cluster #3 (assigned as M2-like macrophages), concatenated CD45+ CD3− events (Conc), and negative control (Neg) relative fluorescence intensity (RFI) values are shown.
Figure 6.
Figure 6.
Increased frequency of circulating exhausted T cells and myeloid-derived suppressor MDSC cells in NRs to nCT prior to treatment. A, Exemplified t-SNE visualization of 17 surface markers representing 32,000 merged CD3+ CD4+ cells PBMCs from CRs (n = 8) and NRs (n = 8) before nCT; 18 clusters were predicted by unsupervised PhenoGraph analysis. B, Histograms showing the expression of six markers in cluster #6 (T effector memory KLRG1high PD-1high), concatenated CD3+ CD4+ events (Conc), and negative control (Neg); relative fluorescence intensity (RFI) values are shown. C, Exemplified t-SNE visualization of 17 surface markers representing 16,000 merged CD3+CD8+ pre-nCT PBMCs from CRs (n = 8) and NRs (n = 8); 14 clusters were predicted by unsupervised PhenoGraph analysis. D, Histograms showing the expression (RFI values) of six markers in cluster #8 (T transitional memory TIGIThigh PD-1high), concatenated CD3+ CD8+ events (Conc), and negative control (Neg). E, Exemplified t-SNE visualization of 20 surface markers representing 15,000 merged CD45+ CD3− pre-nCT PBMCs from CRs (n = 7) and NRs (n = 8). Cells are colored according to the 11 clusters assigned by PhenoGraph and manual annotation; the corresponding heat map shows median marker intensity normalized to a 0 to 1 range. F, Histograms showing the expression (RFI values) of five surface markers in PhenoGraph clusters #1 and #4, concatenated CD45+ CD3− events, and Neg; clusters #1 and #4 were assigned as monocytes HLA-DRhigh (enriched in CRs) and MO-MDSC CCR7+ (enriched in NRs), respectively. G, Frequencies of CD45+CD3− cells in 11 PhenoGraph clusters (determined by 20 surface markers) in CRs (n = 7) and NRs (n = 8), presented as mean ± SEM. **, P < 0.01; one-way ANOVA. H, Correlation between the flow cytometry frequency (%) of CD14+HLA-DRhigh PBMCs and CD39+CD103+CD8+ TILs for n = 12 patients; 95% confidence interval is shaded in gray (R, Spearman correlation; P value).
Figure 7.
Figure 7.
Differential circulating proinflammatory cytokines and complement anaphylatoxins in responders and NRs to nCT. A, Baseline plasma concentrations of GM-CSF, IL4, CXCL10, C3a, and C5a in CRs (n = 12) and NRs(n = 14); bars represent mean ± SEM. *, P < 0.05; **, P < 0.01; unpaired t test. B, Spearman correlation coefficients for the 30 circulating cytokines analyzed and frequency of tumor-infiltrating CD8+ CD39+ CD103+ cells (calculated by flow cytometry manual gating on 15 baseline EACs; *, P < 0.05; **, P < 0.01). C, Correlation between the baseline plasma concentration of RANTES, IL1RA, GM-CSF, and IL4 and flow cytometry frequency (%) of CD39+ CD103+ CD8+ TILs for n = 15 patients; 95% confidence interval (CI) is shaded in gray (R, Spearman correlation; P value). D, Correlation between the baseline plasma concentration of GM-CSF and flow cytometry frequency (%) of circulating CD14+ HLA-DRhigh cells for n = 15 patients; 95% CI is shaded in gray (R, Spearman correlation; P value).

References

    1. Smyth EC, Lagergren J, Fitzgerald RC, Lordick F, Shah MA, Lagergren P, et al. . Oesophageal cancer. Nat Rev Dis Primers 2017;3:17048.
    1. Shapiro J, van Lanschot JJB, Hulshof M, van Hagen P, van Berge Henegouwen MI, Wijnhoven BPL, et al. . Steyerberg EW, van der Gaast A; CROSS study group. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol 2015;16:1090–8.
    1. Obermannová R, Alsina M, Cervantes A, Leong T, Lordick F, Nilsson M, et al. . Oesophageal cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2022;33:992–1004.
    1. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012;12:298–306.
    1. Mennonna D, Maccalli C, Romano MC, Garavaglia C, Capocefalo F, Bordoni R, et al. . T cell neoepitope discovery in colorectal cancer by high throughput profiling of somatic mutations in expressed genes. Gut 2017;66:454–63.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:1350–5.
    1. Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012;11:215–33.
    1. Lhuillier C, Rudqvist NP, Elemento O, Formenti SC, Demaria S. Radiation therapy and anti-tumor immunity: exposing immunogenic mutations to the immune system. Genome Med 2019;11:40.
    1. Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol 2022;23:487–500.
    1. Mandard AM, Dalibard F, Mandard JC, Marnay J, Henry-Amar M, Petiot JF, et al. . Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer 1994;73:2680–6.
    1. Donahue JM, Nichols FC, Li Z, Schomas DA, Allen MS, Cassivi SD, et al. . Complete pathologic response after neoadjuvant chemoradiotherapy for esophageal cancer is associated with enhanced survival. Ann Thorac Surg 2009;87:392–8; discussion 398–9.
    1. Chen Y, Hao Q, Wang J, Li J, Huang C, Zhang Y, et al. . Ubiquitin ligase TRIM71 suppresses ovarian tumorigenesis by degrading mutant p53. Cell Death Dis 2019;10:737.
    1. Jordan MS, Koretzky GA. Coordination of receptor signaling in multiple hematopoietic cell lineages by the adaptor protein SLP-76. Cold Spring Harb Perspect Biol 2010;2:a002501.
    1. Wang Z, Peng M. A novel prognostic biomarker LCP2 correlates with metastatic melanoma-infiltrating CD8+ T cells. Sci Rep 2021;11:9164.
    1. Dollt C, Michel J, Kloss L, Melchers S, Schledzewski K, Becker K, et al. . The novel immunoglobulin super family receptor SLAMF9 identified in TAM of murine and human melanoma influences pro-inflammatory cytokine secretion and migration. Cell Death Dis 2018;9:939.
    1. Wang L, Yin J, Wang X, Shao M, Duan F, Wu W, et al. . C-type lectin-like receptor 2 suppresses AKT signaling and invasive activities of gastric cancer cells by blocking expression of phosphoinositide 3-kinase subunits. Gastroenterology 2016;150:1183–95.
    1. Hu R, Bi R, Jiang L, Yang X, Zhong Y, Xie X. LncRNA TUSC8 suppresses the proliferation and migration of esophageal cancer cells by downregulation of VEGFA. J Cancer 2021;12:6393–400.
    1. Luo Y, Xiang W, Liu Z, Yao L, Tang L, Tan W, et al. . Functional role of the SLC7A11-AS1/xCT axis in the development of gastric cancer cisplatin-resistance by a GSH-dependent mechanism. Free Radic Biol Med 2022;184:53–65.
    1. Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, et al. . Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov 2019;9:1673–85.
    1. Han W, Hu C, Fan ZJ, Shen GL. Transcript levels of keratin 1/5/6/14/15/16/17 as potential prognostic indicators in melanoma patients. Sci Rep. 2021;11:1023.
    1. Iwabu J, Yamashita S, Takeshima H, Kishino T, Takahashi T, Oda I, et al. . FGF5 methylation is a sensitivity marker of esophageal squamous cell carcinoma to definitive chemoradiotherapy. Sci Rep 2019;9:13347.
    1. Toivola DM, Boor P, Alam C, Strnad P. Keratins in health and disease. Curr Opin Cell Biol 2015;32:73–81.
    1. Lee YE, He HL, Shiue YL, Lee SW, Lin LC, Wu TF, et al. . The prognostic impact of lipid biosynthesis-associated markers, HSD17B2 and HMGCS2, in rectal cancer treated with neoadjuvant concurrent chemoradiotherapy. Tumour Biol 2015;36:7675–83.
    1. Luis G, Godfroid A, Nishiumi S, Cimino J, Blacher S, Maquoi E, et al. . Tumor resistance to ferroptosis driven by Stearoyl-CoA Desaturase-1 (SCD1) in cancer cells and Fatty Acid Biding Protein-4 (FABP4) in tumor microenvironment promote tumor recurrence. Redox Biol 2021;43:102006.
    1. Dahabieh MS, Di PE, Jangal M, Goncalves C, Witcher M, Braverman NE, et al. . Peroxisomes and cancer: The role of a metabolic specialist in a disease of aberrant metabolism. Biochim Biophys Acta Rev Cancer 2018;1870:103–21.
    1. Izadi F, Sharpe BP, Breininger SP, Secrier M, Gibson J, Walker RC, et al. . Genomic analysis of response to neoadjuvant chemotherapy in esophageal adenocarcinoma. Cancers (Basel) 2021;13:3394.
    1. Dhatchinamoorthy K, Colbert JD, and Kenneth L. Rock KL. Cancer immune evasion through loss of MHC class I antigen presentation. Front Immunol 2021;12:636568.
    1. McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins TBK, Wilson GA, et al. . Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 2017;171:1259–71.
    1. Cory S, Huang DC, Adams JM. The Bcl-2 family: roles in cell survival and oncogenesis. Oncogene 2003;22:8590–607.
    1. Pearce OMT, Delaine-Smith RM, Maniati E, Nichols S, Wang J, Böhm S, et al. . Deconstruction of a metastatic tumor microenvironment reveals a common matrix response in human cancers. Cancer Discov 2018;8:304–19.
    1. Al-Khyatt W, Tufarelli C, Khan R, Iftikhar SY. Selective oestrogen receptor antagonists inhibit oesophageal cancer cell proliferation in vitro. BMC Cancer 2018;18:121.
    1. Li M, Zhou C. Progesterone receptor gene serves as a prognostic biomarker associated with immune infiltration in gastric cancer: a bioinformatics analysis. Transl Cancer Res 2021;10:2663–77.
    1. Duhen T, Duhen R, Montler R, Moses J, Moudgil T, de Miranda NF, et al. . Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun 2018;9:2724.
    1. Losurdo A, Scirgolea C, Alvisi G, Brummelman J, Errico V, Di Tommaso L, et al. . Single-cell profiling defines the prognostic benefit of CD39high tissue resident memory CD8+ T cells in luminal-like breast cancer. Commun Biol 2021;4:1117.
    1. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol 2019;37:457–95.
    1. Shields JD, Kourtis IC, Tomei AA, Roberts JM, Swartz MA. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science 2010;328:749–52.
    1. Zarei S, Schwenter F, Luy P, Aurrand-Lions M, Morel P, Kopf M, et al. . Role of GM-CSF signaling in cell-based tumor immunization. Blood 2009;113:6658–68.
    1. Mandruzzato S, Solito S, Falisi E, Francescato S, Chiarion-Sileni V, Mocellin S, et al. . IL4Ralpha+ myeloid-derived suppressor cell expansion in cancer patients. J Immunol 2009;182:6562–8.
    1. Liu H, Ling CC, Yeung WHO, Pang L, Liu J, Zhou J, et al. . Monocytic MDSC mobilization promotes tumor recurrence after liver transplantation via CXCL10/TLR4/MMP14 signaling. Cell Death Dis 2021;12:489.
    1. Llaudo I, Fribourg M, Medof ME, Conde P, Ochando J, Heeger PS. C5aR1 regulates migration of suppressive myeloid cells required for costimulatory blockade-induced murine allograft survival. Am J Transplant 2019;19:633–45.
    1. Turkington RC, Knight LA, Blayney JK, Secrier M, Douglas R, Parkes EE, et al. . Immune activation by DNA damage predicts response to chemotherapy and survival in oesophageal adenocarcinoma. Gut 2019;68:1918–27.
    1. Goedegebuure RSA, Harrasser M, de Klerk LK, van Schooten TS, van Grieken NCT, Eken M, et al. . Pre-treatment tumor-infiltrating T cells influence response to neoadjuvant chemoradiotherapy in esophageal adenocarcinoma. Oncoimmunology 2021;10:1954807.
    1. Soeratram TT, Creemers A, Meijer SL, de Boer OJ, Vos W, Hooijer GK, et al. . Tumor-immune landscape patterns before and after chemoradiation in resectable esophageal adenocarcinomas. J Pathol 2022;256:282–96.
    1. Tian T, Li Z. Targeting Tim-3 in cancer with resistance to PD-1/PD-L1 blockade. Front Oncol 2021;11:731175.
    1. Cerboni S, Jeremiah N, Gentili M, Gehrmann U, Conrad C, Stolzenberg MC, et al. . Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes. J Exp Med 2017;214:1769–85.
    1. Oshi M, Takahashi H, Tokumaru Y, Yan L, Rashid OM, Nagahashi M, et al. . The E2F pathway score as a predictive biomarker of response to neoadjuvant therapy in ER+/HER2- breast cancer. Cells 2020;9:1643.
    1. Bacci M, Lorito N, Smiriglia A, Morandi A. Fat and furious: lipid metabolism in antitumoral therapy response and resistance. Trends Cancer 2021;7:198–213.
    1. Leone RD, Powell JD. Metabolism of immune cells in cancer. Nat Rev Cancer 2020;20:516–31.
    1. Arner EN, Rathmell JC. Metabolic programming and immune suppression in the tumor microenvironment. Cancer Cell 2023;41:421–33.
    1. Pedrosa L, Foguet C, Oliveres H, Archilla I, de Herreros MG, Rodríguez A, et al. . A novel gene signature unveils three distinct immune-metabolic rewiring patterns conserved across diverse tumor types and associated with outcomes. Front Immunol 2022;13:926304.
    1. Ma X, Bi E, Lu Y, Su P, Huang C, Liu L, et al. . Cholesterol induces CD8+ T cell exhaustion in the tumor microenvironment. Cell Metab 2019;30:143–56.
    1. Harding JJ, Moreno V, Bang YJ, Hong MH, Patnaik A, Trigo J, et al. . Blocking TIM-3 in treatment-refractory advanced solid tumors: a phase Ia/b study of LY3321367 with or without an Anti-PD-L1 antibody. Clin Cancer Res 2021;27:2168–78.
    1. Kelly RJ, Ajani JA, Kuzdzal J, Zander T, Van Cutsem E, Piessen G, et al. . Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer. N Engl J Med 2021;384:1191–203.
    1. van den Ende T, de Clercq NC, van Berge Henegouwen MI, Gisbertz SS, Geijsen ED, Verhoeven RHA, et al. . Neoadjuvant chemoradiotherapy combined with atezolizumab for resectable esophageal adenocarcinoma: a single-arm phase II feasibility trial (PERFECT). Clin Cancer Res 2021;27:3351–9.

Source: PubMed

3
Abonnere