Clinical Positioning of the IAP Antagonist Tolinapant (ASTX660) in Colorectal Cancer

Nyree Crawford, Katie J Stott, Tamas Sessler, Christopher McCann, William McDaid, Andrea Lees, Cheryl Latimer, Jennifer P Fox, Joanne M Munck, Tomoko Smyth, Alpesh Shah, Vanessa Martins, Mark Lawler, Philip D Dunne, Emma M Kerr, Simon S McDade, Vicky M Coyle, Daniel B Longley, Nyree Crawford, Katie J Stott, Tamas Sessler, Christopher McCann, William McDaid, Andrea Lees, Cheryl Latimer, Jennifer P Fox, Joanne M Munck, Tomoko Smyth, Alpesh Shah, Vanessa Martins, Mark Lawler, Philip D Dunne, Emma M Kerr, Simon S McDade, Vicky M Coyle, Daniel B Longley

Abstract

Inhibitors of apoptosis proteins (IAPs) are intracellular proteins, with important roles in regulating cell death, inflammation, and immunity. Here, we examined the clinical and therapeutic relevance of IAPs in colorectal cancer. We found that elevated expression of cIAP1 and cIAP2 (but not XIAP) significantly correlated with poor prognosis in patients with microsatellite stable (MSS) stage III colorectal cancer treated with 5-fluorouracil (5FU)-based adjuvant chemotherapy, suggesting their involvement in promoting chemoresistance. A novel IAP antagonist tolinapant (ASTX660) potently and rapidly downregulated cIAP1 in colorectal cancer models, demonstrating its robust on-target efficacy. In cells co-cultured with TNFα to mimic an inflammatory tumor microenvironment, tolinapant induced caspase-8-dependent apoptosis in colorectal cancer cell line models; however, the extent of apoptosis was limited because of inhibition by the caspase-8 paralogs FLIP and, unexpectedly, caspase-10. Importantly, tolinapant-induced apoptosis was augmented by FOLFOX in human colorectal cancer and murine organoid models in vitro and in vivo, due (at least in part) to FOLFOX-induced downregulation of class I histone deacetylases (HDAC), leading to acetylation of the FLIP-binding partner Ku70 and downregulation of FLIP. Moreover, the effects of FOLFOX could be phenocopied using the clinically relevant class I HDAC inhibitor, entinostat, which also induced acetylation of Ku70 and FLIP downregulation. Further analyses revealed that caspase-8 knockout RIPK3-positive colorectal cancer models were sensitive to tolinapant-induced necroptosis, an effect that could be exploited in caspase-8-proficient models using the clinically relevant caspase inhibitor emricasan. Our study provides evidence for immediate clinical exploration of tolinapant in combination with FOLFOX in poor prognosis MSS colorectal cancer with elevated cIAP1/2 expression.

Trial registration: ClinicalTrials.gov NCT03871959.

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

Figures

Figure 1.
Figure 1.
Correlation between high cIAP1/2 expression and prognosis. Analysis of patients with stage III MSS who received adjuvant fluorouracil-based treatment extracted from GSE39582 cohort. A, Kaplan–Meier plots of 5-year OS. Graphs comparing the difference in survival probability based on BIRC2 and BIRC3 expression levels alone. Results are shown with the log-rank test, the HR with its 95% confidential interval and the Wald test of significance. B–E, Difference of pathway activation by ssGSEA (B) and cell type profiling (D) by xCELL between cIAP1 or cIAP2 high and low expressing patients was assessed by one-way ANOVA model. Numbers shown are the calculated P values. Heatmap were drawn showing the Z-scores of the significantly different ssGSEA pathways (C) and immune or stroma infiltration by xCELL (E).
Figure 2.
Figure 2.
Sensitivity of colorectal cancer cell lines to IAP inhibitor, tolinapant. A, Cell viability assay in 36 colorectal cancer cell line panel following 72 hours treatment with tolinapant (tolin; 10-point assay 0.0005–10 μmol/L) alone and in combination with TNFα (1 ng/mL), represented as relative activity area (%), calculated as (experimental AUC/max AUC possible). Cell lines categorized as sensitive to tolinapant/TNFα are highlighted in green, intermediate in white, and resistant in orange. B, Western blot analysis of basal expression of FLIP(L), FLIP(S), Mcl-1, cIAP1, cIAP2, XIAP, BAX, SMAC, RIPK1, FADD, procaspase-8, procaspase-10, RIPK3 and MLKL and β-actin in a panel of colorectal cancer cell lines. C, Cell surface expression of TNFR1 in DLD1, SW620, LoVo, HT29, and HCT116 cell lines, represented as % of TNFR1-positive cells compared with an IgG isotype control. D, Western blot analysis of cIAP1, cIAP2, XIAP, and β-actin in HCT116, HT29, and DLD-1 cells following treatment with 1 μmol/L, 100 nmol/L, 10 nmol/L, 1 nmol/L, and 0.1 nmol/L tolinapant for 24 hours. E, Western blot analysis of RIPK1 and procaspase-8 following immunoprecipitation of caspase-8 from HCT116, HT29, and DLD-1 cells treated with 1 ng/mL TNFα, 1 μmol/L tolinapant and a combination of TNFα/tolinapant for 3 hours in the presence of 20 μmol/L z-VAD-fmk. An IgG isotype control was used as a control. F, Annexin V/PI analysis in HCT116, HT29, and DLD-1 cells following treatment with 1 ng/mL TNFα, 1 μmol/L tolinapant, or a combination of TNFα/tolinapant for 24 hours. G, Tumor volume (mm3) in HCT116, HT29, and DLD-1 xenograft models treated with vehicle and 16 mg/kg tolinapant. Data show mean tumor volume per treatment group per timepoint ± SEM (HCT116; vehicle n = 10, tolinapant n = 10, HT29; vehicle n = 7, tolinapant n = 7, SW620; vehicle n = 10, tolinapant n = 12). Results were compared using a two-tailed Student t test, *, P < 0.05; **, P < 0.01; and ***, P < 0.001
Figure 3.
Figure 3.
Tolinapant synergizes with chemotherapy. A, Annexin V/PI analysis in HCT116, SW620, and HT29 cells treated with 1 ng/mL TNFα, 1 μmol/L tolinapant, or a combination of tolinapant/TNFα in the presence and absence of 5 μmol/L 5FU and 1 μmol/L oxaliplatin (OX) for 72 hours. B, Western blot analysis of PARP, cIAP2, cIAP1, XIAP, FLIP(L), FLIP(S), and β-actin in HCT116, SW620, and HT29 cells treated with 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FOLFOX), 1 μmol/L tolinapant and a combination of FOLFOX/tolinapant (COMBO) for 24 and 48 hours. C, Pictures from AKP and AK organoids treated with 1 μmol/L tolinapant/1 ng/mL TNFα (tolin/TNF), 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FF) or combination (COMBO) for 72 hours. Diameter in AU from AKP (n = 75 for each treatment group) and AK (n = 75 for each treatment group) organoids treated with 1 μmol/L tolinapant/1 ng/mL TNFα (tolin/TNF), 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FOLFOX) or combination (COMBO) for 72 hours. D, Cell viability assays in AKP and AK organoids treated with 1 μmol/L tolinapant/1 ng/mL TNFα (tolin/TNF), 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FOLFOX) or combination (COMBO) for 72 hours. E, Western blot analysis of basal expression of cIAP1, p53, and β-actin in AK and AKP organoids. Western blot analysis of cIAP1 and β-actin in AK and AKP organoids treated with 1 μmol/L tolinapant for 24 hours. F, Tumor volume (mm3) of AKP organoids transplanted into C57BL/6 mice treated with vehicle (n = 3), 5FU (10 mg/kg) + oxaliplatin (1 mg/kg; FOLFOX; n = 4), tolinapant (16 mg/kg; n = 4) and combination (n = 4). Data show mean tumor volume per treatment group per timepoint ± SEM. G, Densitometry from Western blot analysis of cIAP1 normalized to β-actin in AKP organoids transplanted into C57BL/6 mice and treated with vehicle (n = 3), 5FU (10 mg/kg) + oxaliplatin (1 mg/kg; FOLFOX; n = 4), tolinapant (16 mg/kg; n = 4), and combination (n = 3). Results were compared using a two-tailed Student t test, *, P < 0.05; **, P < 0.01; and ***, P < 0.001.
Figure 4.
Figure 4.
Role of FLIP in determining sensitivity to tolinapant. A, Expression of caspase-8 and FLIP mRNA [log2 (TPM (transcripts per million) + 1] in colorectal cancer cell lines categorized as sensitive or resistant to tolinapant/TNFα in the cell line screen in Fig. 2A. B, Annexin V/PI analysis in HCT116, SW620, HT29, DLD-1, and LoVo cells transfected with control (SCR) and FLIP(T) siRNA for 24 hours prior to treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα for a further 24 hours. C, Cell viability assays in HCT116, SW620, HT29, DLD-1, and LoVo cells transfected with control (SCR) and FLIP(T) siRNA for 6 hours prior to treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα for a further 24 hours. D, Western blot analysis of PARP, FLIP(L), FLIP(S), cIAP1, and β-actin and in HCT116, SW620, and HT29 cells transfected with control (SCR) and FLIP(T) siRNA for 24 hours prior to treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα for a further 24 hours. Results were compared using a two-tailed Student t test, *, P < 0.05; **, P < 0.01; and ***, P < 0.001.
Figure 5.
Figure 5.
Mechanism of chemotherapy-induced sensitization to tolinapant. A, Western blot analysis of FLIP(L), FLIP(S), and β-actin in HCT116 cells retrovirally overexpressing pbabe empty vector (EV), pbabe FLIP(S), pbabe FLIP(S) F114A, and pbabe FLIP(L). B, Annexin V/PI analysis of in HCT116 cells retrovirally overexpressing pbabe empty vector (EV), pbabe FLIP(S), pbabe FLIP(S) F114A, and pbabe FLIP(L) following treatment with 1 μmol/L tolinapant/1 ng/mL TNFα, 5 μmol/L 5FU, and 1 μmol/L oxaliplatin (FOLFOX) or combination for 48 hours. C, Western blot analysis of PARP, cIAP1, FLIP(L), FLIP(S), procaspase-8, procaspase-10, and β-actin in HCT116 WT and HCT116 caspase-8 KO cells treated with 1 μmol/L tolinapant/1 ng/mL TNFα (tolin/TNFα), 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FF) or combination for 48 hours. D, Annexin V/PI analysis in HCT116 WT and HCT116 caspase-8 KO cells treated with 1 μmol/L tolinapant/1 ng/mL TNFα, 5 μmol/L 5FU, and 1 μmol/L oxaliplatin (FOLFOX) or combination for 72 hours. E, Western blot analysis of procaspase-10, PARP, cleaved caspase-3, FLIP(L), FLIP(S), cIAP1, and β-actin in HCT116 and DLD1 cells transfected with control (SCR) and caspase-10 (siC10) siRNA for 24 hours prior to treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα for a further 24 hours. F, Caspase-3/7 activity assay in HCT116 cells transfected with control (SCR) and caspase-10 siRNA (siC10) for 24 hours prior to treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα (tolin/TNFα) for a further 24 hours. G, Western blot analysis of procaspase-10, procaspase-8, and β-actin in HCT116 WT, HCT116 caspase-8 KO, HCT116 caspase-10 KO and HCT116 caspase-8/caspase-10 DKO cells, and Annexin V/PI analysis in HCT116 WT and HCT116 caspase-8 KO, HCT116 caspase-10 KO and HCT116 caspase-8/caspase-10 DKO cells treated with 1 μmol/L tolinapant/1 ng/mL TNFα, 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FOLFOX) or combination for 48 hours. H, Western blot analysis of HDAC1, HDAC2, HDAC3, and β-actin in HCT116 WT and HCT116 caspase-8 KO cells treated with 1 μmol/L tolinapant/1 ng/mL TNFα (tolin/TNFα), 5 μmol/L 5FU, and 1 μmol/L oxaliplatin (FF) or combination for 48 hours. I, Western blot analysis of Ku70 following immunoprecipitation of acetylated lysine (AcK) from HCT116 cells treated with 5 μmol/L 5FU and 1 μmol/L oxaliplatin (FOLFOX) for 24 hours. An IgG isotype control was used as a control. J, Western blot and caspase-3/7 activity analyses of Ku70, FLIP(L), FLIP(S), procaspase-10, PARP, cIAP1, CIAP2, XIAP, and β-actin and in HCT116 cells transfected with control (SCR) and Ku70 siRNA (siKU70) for 24 hours prior to treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα (tolin/TNFα) for a further 24 hours.
Figure 6.
Figure 6.
Necroptotic cell death induced by tolinapant in RIPK3-expressing colorectal cancer cells. A, Cell viability assays in HT29 caspase-8 WT and HT29 caspase-8 KO cells pretreated with 2.5 μmol/L entinostat for 24 hours then addition of 1 μmol/L tolinapant and 1 ng/mL TNFα for a further 24 hours. B, Western blot analysis of PARP, p-MLKL(Ser358), total MLKL, p-RIPK3(Ser227), total RIPK3, p-RIPK1(Ser166), total RIPK1, cIAP1, caspase-8, and β-actin in HT29 caspase-8 WT and HT29 caspase-8 KO cells 24 hours following treatment with 1 μmol/L tolinapant and 1 ng/mL TNFα. C, PI analysis in HT29 caspase-8 WT and HT29 caspase-8 KO cells pretreated for 30 minutes with 10 μmol/L z-VAD and 2 μmol/L necrosulfonamide then addition of 1 μmol/L tolinapant and 1 ng/mL TNFα for 24 hours. D, Annexin V/PI analysis in HT29 cells pretreated with 10 μmol/L z-VAD, 5 μmol/L emricasan or 10 μmol/L emricasan for 30 minutes then addition of 1 μmol/L tolinapant and 1 ng/mL TNFα for 24 hours. E, Western blot analysis of p-MLKL(Ser358), total MLKL, p-RIPK3(Ser227), total RIPK3, p-RIPK1 (Ser166), total RIPK1, cIAP1, and β-actin in HT29 cells pretreated with 10 μmol/L emricasan for 30 minutes then addition of 1 μmol/L tolinapant and 1 ng/mL TNFα for 24 hours. F, Pictures from AK and AKP organoids pretreated with 10 μmol/L emricasan for 30 minutes then addition of 1 μmol/L tolinapant and 1 ng/mL TNFα for 48 hours. G, Western blot analysis of p-RIPK3 (Thr231/Ser232), total RIPK3, cIAP1, and β-actin in AKP organoids pretreated with 10 μmol/L necrostatin-1 and 10 μmol/L emricasan (Emri) for 30 minutes before the addition of 1 μmol/L tolinapant and 1 ng/mL TNFα (tolin/TNF) for 24 hours. Results were compared using a two-tailed Student t test, *, P < 0.05; **, P < 0.01; and ***, P < 0.001.

References

    1. Van Schaeybroeck S, Allen WL, Turkington RC, Johnston PG. Implementing prognostic and predictive biomarkers in CRC clinical trials. Nat Rev Clin Oncol 2011;8:222–32.
    1. Quasar Collaborative G, Gray R, Barnwell J, McConkey C, Hills RK, Williams NS, et al. . Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet 2007;370:2020–9.
    1. Efficacy of adjuvant fluorouracil and folinic acid in colon cancer. International Multicentre Pooled Analysis of Colon Cancer Trials (IMPACT) investigators. Lancet 1995;345:939–44.
    1. Suzuki Y, Nakabayashi Y, Takahashi R. Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its anti-apoptotic effect in Fas-induced cell death. Proc Natl Acad Sci U S A 2001;98:8662–7.
    1. Bertrand MJ, Milutinovic S, Dickson KM, Ho WC, Boudreault A, Durkin J, et al. . cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol Cell 2008;30:689–700.
    1. Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 2003;114:181–90.
    1. Sun XS, Tao Y, Le Tourneau C, Pointreau Y, Sire C, Kaminsky MC, et al. . Debio 1143 and high-dose cisplatin chemoradiotherapy in high-risk locoregionally advanced squamous cell carcinoma of the head and neck: a double-blind, multicentre, randomised, phase 2 study. Lancet Oncol 2020;21:1173–87.
    1. Johnson CN, Ahn JS, Buck IM, Chiarparin E, Day JEH, Hopkins A, et al. . A fragment-derived clinical candidate for antagonism of X-linked and cellular inhibitor of apoptosis proteins: 1-(6-[(4-Fluorophenyl)methyl]-5-(hydroxymethyl)-3,3-dimethyl-1 H, 2 H, 3 H-pyrrolo[3, 2- b]pyridin-1-yl)-2-[(2 R, 5 R)-5-methyl-2-([(3R)-3-methylmorpholin-4-yl]methyl)piperazin-1-yl]ethan-1-one (ASTX660). J Med Chem 2018;61:7314–29.
    1. Mita MM, LoRusso PM, Papadopoulos KP, Gordon MS, Mita AC, Ferraldeschi R, et al. . A phase 1 study of ASTX660, an antagonist of inhibitors of apoptosis proteins, in adults with advanced cancers or lymphoma. Clin Cancer Res 2020;26:2819–26.
    1. Marisa L, de Reynies A, Duval A, Selves J, Gaub MP, Vescovo L, et al. . Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value. PLoS Med 2013;10:e1001453.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 2008;9:559.
    1. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol 2017;18:220.
    1. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet 2006;38:500–1.
    1. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25.
    1. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. . The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 2012;483:603–7.
    1. Schwitalla S, Fingerle AA, Cammareri P, Nebelsiek T, Goktuna SI, Ziegler PK, et al. . Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell 2013;152:25–38.
    1. Cammareri P, Vincent DF, Hodder MC, Ridgway RA, Murgia C, Nobis M, et al. . TGFbeta pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis. Cell Death Differ 2017;24:1681–93.
    1. Crawford N, Stasik I, Holohan C, Majkut J, McGrath M, Johnston PG, et al. . SAHA overcomes FLIP-mediated inhibition of SMAC mimetic-induced apoptosis in mesothelioma. Cell Death Dis 2013;4:e733.
    1. Kerr E, Holohan C, McLaughlin KM, Majkut J, Dolan S, Redmond K, et al. . Identification of an acetylation-dependant Ku70/FLIP complex that regulates FLIP expression and HDAC inhibitor-induced apoptosis. Cell Death Differ 2012;19:1317–27.
    1. Lees A, McIntyre AJ, Crawford NT, Falcone F, McCann C, Holohan C, et al. . The pseudo-caspase FLIP(L) regulates cell fate following p53 activation. Proc Natl Acad Sci U S A 2020;117:17808–19.
    1. McCann C, Crawford N, Majkut J, Holohan C, Armstrong CWD, Maxwell PJ, et al. . Cytoplasmic FLIP(S) and nuclear FLIP(L) mediate resistance of castrate-resistant prostate cancer to apoptosis induced by IAP antagonists. Cell Death Dis 2018;9:1081.
    1. Parameswaran N, Patial S. Tumor necrosis factor-alpha signaling in macrophages. Crit Rev Eukaryot Gene Expr 2010;20:87–103.
    1. Varfolomeev E, Blankenship JW, Wayson SM, Fedorova AV, Kayagaki N, Garg P, et al. . IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis. Cell 2007;131:669–81.
    1. Feltham R, Bettjeman B, Budhidarmo R, Mace PD, Shirley S, Condon SM, et al. . Smac mimetics activate the E3 ligase activity of cIAP1 protein by promoting RING domain dimerization. J Biol Chem 2011;286:17015–28.
    1. Wong WW, Vince JE, Lalaoui N, Lawlor KE, Chau D, Bankovacki A, et al. . cIAPs and XIAP regulate myelopoiesis through cytokine production in an RIPK1- and RIPK3-dependent manner. Blood 2014;123:2562–72.
    1. Majkut J, Sgobba M, Holohan C, Crawford N, Logan AE, Kerr E, et al. . Differential affinity of FLIP and procaspase 8 for FADD's DED binding surfaces regulates DISC assembly. Nat Commun 2014;5:3350.
    1. Humphreys LM, Fox JP, Higgins CA, Majkut J, Sessler T, McLaughlin K, et al. . A revised model of TRAIL-R2 DISC assembly explains how FLIP(L) can inhibit or promote apoptosis. EMBO Rep 2020;21:e49254.
    1. Muhlethaler-Mottet A, Flahaut M, Bourloud KB, Nardou K, Coulon A, Liberman J, et al. . Individual caspase-10 isoforms play distinct and opposing roles in the initiation of death receptor-mediated tumour cell apoptosis. Cell Death Dis 2011;2:e125.
    1. Horn S, Hughes MA, Schilling R, Sticht C, Tenev T, Ploesser M, et al. . Caspase-10 negatively regulates Caspase-8-mediated cell death, switching the response to CD95L in favor of NF-kappaB activation and cell survival. Cell Rep 2017;19:785–97.
    1. Shiffman M, Freilich B, Vuppalanchi R, Watt K, Chan JL, Spada A, et al. . Randomised clinical trial: emricasan versus placebo significantly decreases ALT and caspase 3/7 activation in subjects with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2019;49:64–73.
    1. Krajewska M, Kim H, Kim C, Kang H, Welsh K, Matsuzawa S, et al. . Analysis of apoptosis protein expression in early-stage colorectal cancer suggests opportunities for new prognostic biomarkers. Clin Cancer Res 2005;11:5451–61.
    1. Flanagan L, Kehoe J, Fay J, Bacon O, Lindner AU, Kay EW, et al. . High levels of X-linked Inhibitor-of-Apoptosis Protein (XIAP) are indicative of radio chemotherapy resistance in rectal cancer. Radiat Oncol 2015;10:131.
    1. Xiao R, Allen CT, Tran L, Patel P, Park SJ, Chen Z, et al. . Antagonist of cIAP1/2 and XIAP enhances anti-tumor immunity when combined with radiation and PD-1 blockade in a syngeneic model of head and neck cancer. Oncoimmunology 2018;7:e1471440.
    1. Xiao R, An Y, Ye W, Derakhshan A, Cheng H, Yang X, et al. . Dual antagonist of cIAP/XIAP ASTX660 sensitizes HPV(-) and HPV(+) head and neck cancers to TNFalpha, TRAIL, and radiation therapy. Clin Cancer Res 2019;25:6463–74.
    1. Mlecnik B, Bindea G, Angell HK, Maby P, Angelova M, Tougeron D, et al. . Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity 2016;44:698–711.
    1. Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS Jr. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 1998;281:1680–3.
    1. Feoktistova M, Geserick P, Kellert B, Dimitrova DP, Langlais C, Hupe M, et al. . cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol Cell 2011;43:449–63.
    1. Lin Y, Devin A, Rodriguez Y, Liu ZG. Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Genes Dev 1999;13:2514–26.
    1. Qin J, Wen B, Liang Y, Yu W, Li H. Histone modifications and their role in colorectal cancer (Review). Pathol Oncol Res 2020;26:2023–33.
    1. Featherstone C, Jackson SP. Ku, a DNA repair protein with multiple cellular functions? Mutat Res 1999;434:3–15.
    1. Chen M, Orozco A, Spencer DM, Wang J. Activation of initiator caspases through a stable dimeric intermediate. J Biol Chem 2002;277:50761–7.
    1. He S, Wang L, Miao L, Wang T, Du F, Zhao L, et al. . Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 2009;137:1100–11.
    1. Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, et al. . Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol 2014;16:55–65.
    1. Aaes TL, Kaczmarek A, Delvaeye T, De Craene B, De Koker S, Heyndrickx L, et al. . Vaccination with necroptotic cancer cells induces efficient anti-tumor immunity. Cell Rep 2016;15:274–87.
    1. Yatim N, Jusforgues-Saklani H, Orozco S, Schulz O, Barreira da Silva R, Reis e Sousa C, et al. . RIPK1 and NF-kappaB signaling in dying cells determines cross-priming of CD8(+) T cells. Science 2015;350:328–34.
    1. Moriwaki K, Bertin J, Gough PJ, Orlowski GM, Chan FK. Differential roles of RIPK1 and RIPK3 in TNF-induced necroptosis and chemotherapeutic agent-induced cell death. Cell Death Dis 2015;6:e1636.
    1. Conev NV, Dimitrova EG, Bogdanova MK, Kashlov YK, Chaushev BG, Radanova MA, et al. . RIPK3 expression as a potential predictive and prognostic marker in metastatic colon cancer. Clin Invest Med 2019;42:E31–E38.
    1. Najafov A, Zervantonakis IK, Mookhtiar AK, Greninger P, March RJ, Egan RK, et al. . BRAF and AXL oncogenes drive RIPK3 expression loss in cancer. PLoS Biol 2018;16:e2005756.
    1. Brumatti G, Ma C, Lalaoui N, Nguyen NY, Navarro M, Tanzer MC, et al. . The caspase-8 inhibitor emricasan combines with the SMAC mimetic birinapant to induce necroptosis and treat acute myeloid leukemia. Sci Transl Med 2016;8:339ra369.
    1. Ye W, Gunti S, Allen CT, Hong Y, Clavijo PE, Van Waes C, et al. . ASTX660, an antagonist of cIAP1/2 and XIAP, increases antigen processing machinery and can enhance radiation-induced immunogenic cell death in preclinical models of head and neck cancer. Oncoimmunology 2020;9:1710398.

Source: PubMed

3
S'abonner