Identification of TRAIL-inducing compounds highlights small molecule ONC201/TIC10 as a unique anti-cancer agent that activates the TRAIL pathway

Joshua E Allen, Gabriel Krigsfeld, Luv Patel, Patrick A Mayes, David T Dicker, Gen Sheng Wu, Wafik S El-Deiry, Joshua E Allen, Gabriel Krigsfeld, Luv Patel, Patrick A Mayes, David T Dicker, Gen Sheng Wu, Wafik S El-Deiry

Abstract

Background: We previously reported the identification of ONC201/TIC10, a novel small molecule inducer of the human TRAIL gene that improves efficacy-limiting properties of recombinant TRAIL and is in clinical trials in advanced cancers based on its promising safety and antitumor efficacy in several preclinical models.

Methods: We performed a high throughput luciferase reporter screen using the NCI Diversity Set II to identify TRAIL-inducing compounds.

Results: Small molecule-mediated induction of TRAIL reporter activity was relatively modest and the majority of the hit compounds induced low levels of TRAIL upregulation. Among the candidate TRAIL-inducing compounds, TIC9 and ONC201/TIC10 induced sustained TRAIL upregulation and apoptosis in tumor cells in vitro and in vivo. However, ONC201/TIC10 potentiated tumor cell death while sparing normal cells, unlike TIC9, and lacked genotoxicity in normal fibroblasts. Investigating the effects of TRAIL-inducing compounds on cell signaling pathways revealed that TIC9 and ONC201/TIC10, which are the most potent inducers of cell death, exclusively activate Foxo3a through inactivation of Akt/ERK to upregulate TRAIL and its pro-apoptotic death receptor DR5.

Conclusion: These studies reveal the selective activity of ONC201/TIC10 that led to its selection as a lead compound for this novel class of antitumor agents and suggest that ONC201/TIC10 is a unique inducer of the TRAIL pathway through its concomitant regulation of the TRAIL ligand and its death receptor DR5.

Figures

Figure 1
Figure 1
Identification of TRAIL-inducing compounds by high-throughput luciferase reporter screen. (a) Plot of reporter induction versus signal for screen conducted with the NCI Diversity Set II at 1 μM in HCT116 Bax−/− cells that stably express a luciferase gene reporter construct under transcriptional control of the first 504 base pairs of the human TRAIL gene promoter. Induction is expressed as luminescence relative to DMSO-treated cells. The horizontal red line represents the cutoff for selection. (b) Validation of reporter induction for candidate screen hits in HCT116 Bax−/− cells at multiple doses and time points. Reporter signal is expressed relative to DMSO-treated cells and normalized to cell viability. The horizontal red line represents the cutoff for selection. Data represent an average of three replicates. The horizontal red line represents the cutoff for selection. (C) Molecular structures of the selected TRAIL-inducing compounds.
Figure 2
Figure 2
TIC9 and ONC201/TIC10 induce TRAIL gene transcription and surface TRAIL upregulation in tumor cells in vitro. (a) Reporter activity in HCT116 Bax−/− cells that stably express a luciferase gene reporter construct under transcriptional control of the first 504 base pairs of the human TRAIL gene promoter (n = 3). Reporter signal is expressed relative to DMSO-treated cells and normalized to cell viability. (b) RT-qPCR analysis of TRAIL gene transcription in response to TRAIL-inducing compounds (TICs) (5 μM) in HCT116 p53−/− cells (n = 4, 48 hr). (c) Flow cytometry analysis of surface TRAIL in response to TICs (5 μM) in HCT116 p53−/− cells (n = 3, 72 hr). *P < .05 by student’s two-tailed t test. Error bars represent standard deviation.
Figure 3
Figure 3
TIC4, TIC9, and ONC201/TIC10 upregulate TRAIL in vivo. (a) RT-qPCR analysis of HCT116 p53−/− tumor xenograft in athymic nude mice harvested 2 days following a single intravenous dose of DMSO, TIC4, TIC9, or ONC201/TIC10 (25 mg/kg). *P < .05 by student’s two-tailed t test. (b) IHC analysis of TRAIL protein levels in HCT116 p53−/− tumor xenograft in athymic nude mice harvested following a single intravenous dose of DMSO, TIC4, TIC9, or ONC201/TIC10 (25 mg/kg). (c) Exemplary images and (d) quantification of TUNEL staining in TIC-treated xenograft tumors described in (a). (e) Bioluminescent imaging of luciferase-infected HCT116 subcutaneous xenografts following a single intraperitoneal dose of TIC9 and ONC201/TIC10 (100 mg/kg) on day 0. Quantification of tumor signals shown in right panel (n = 6). Error bars represent standard deviation.
Figure 4
Figure 4
TIC9 and ONC201/TIC10 induce tumor cell death and ONC201/TIC10-induced cell death is tumor-selective. (a) Sub-G1 DNA content of HCT116 p53−/− cells treated with TICs for indicated time periods (1 μM, n = 3). (b) Cell cycle profiles of HCT116 p53−/− cells treated with TICs (5 μM, 72 hr). (c) Sub-G1 DNA content of HCT116 p53−/− cells treated with TICs (72 hrs, 5 μM, n = 3). (d) Sub-G1 DNA content of HFF normal human fibroblasts treated with TICs (72 hr, 5 μM, n = 3). (e) Cell viability assay of HCT116 or HFF cells treated with ONC201/TIC10 at the indicated doses (72 hr, n = 3). Error bars represent standard deviation.
Figure 5
Figure 5
ONC201/TIC10 is not genotoxic to normal human fibroblasts. (a) Bright field microscopy images of HFF fibroblasts following 72 hr incubation with DMSO, ONC201/TIC10, or doxorubicin (1 μM) as indicated. (b) Immunofluorescence imaging of gamma H2AX (green) and nuclear counterstain (blue) in HFF fibroblasts treated as described in (a).
Figure 6
Figure 6
Selection of ONC201/TIC10 as a lead TRAIL-inducing compound. Prioritization assays are shown for the process of selecting ONC201/TIC10 as a lead compound.
Figure 7
Figure 7
ONC201/TIC10 and TIC9 exclusively affect Akt/ERK/Foxo3a. Western blot analysis of HCT116 p53−/− cells treated with TICs in HCT116 p53−/− cells (5 μM, 60 hr).

References

    1. Ashkenazi A. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer. 2002;2:420–30. doi: 10.1038/nrc821.
    1. Wiley SR, Schooley K, Smolak PJ, Din WS, Huang CP, Nicholl JK, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 1995;3:673–82. doi: 10.1016/1074-7613(95)90057-8.
    1. Fanger NA, Maliszewski CR, Schooley K, Griffith TS. Human dendritic cells mediate cellular apoptosis via tumor necrosis factor-related apoptosis-inducing ligand (Trail) J Exp Med. 1999;190:1155–64. doi: 10.1084/jem.190.8.1155.
    1. Griffith TS, Wiley SR, Kubin MZ, Sedger LM, Maliszewski CR, Fange NA. Monocyte-mediated tumoricidal activity via the tumor necrosis factor-related cytokine, TRAIL. J Exp Med. 1999;189:1343–54. doi: 10.1084/jem.189.8.1343.
    1. Kemp TJ, Moore JM, Griffith TS. Human B cells express functional TRAIL/Apo-2 ligand after CpG-containing oligodeoxynucleotide stimulation. J Immunol. 2004;173:892–9. doi: 10.4049/jimmunol.173.2.892.
    1. Zamai L, Ahmad M, Bennett IM, Azzoni L, Alnemri ES, Perussia B. Natural Killer (NK) cell-mediated cytotoxicity: differential use of TRAIL and fas ligand by immature and mature primary human NK cells. J Exp Med. 1998;188:2375–80. doi: 10.1084/jem.188.12.2375.
    1. Greco FA, Bonomi P, Crawford J, Kelly K, Oh Y, Halpern W, et al. Phase 2 study of mapatumumab, a fully human agonistic monoclonal antibody which targets and activates the TRAIL receptor-1, in patients with advanced non-small cell lung cancer. Lung Cancer. 2008;61:82–90. doi: 10.1016/j.lungcan.2007.12.011.
    1. Camidge DR, Herbst RS, Gordon MS, Eckhardt SG, Kurzrock R, Durbin B, et al. A phase I safety and pharmacokinetic study of the death receptor 5 agonistic antibody PRO95780 in patients with advanced malignancies. Clin Cancer Res. 2010;16:1256–63. doi: 10.1158/1078-0432.CCR-09-1267.
    1. Trarbach T, Moehler M, Heinemann V, Köhne CH, Przyborek M, Schulz C, et al. Phase II trial of mapatumumab, a fully human agonistic monoclonal antibody that targets and activates the tumour necrosis factor apoptosis-inducing ligand receptor-1 (TRAIL-R1), in patients with refractory colorectal cancer. Br J Cancer. 2010;102:506–12. doi: 10.1038/sj.bjc.6605507.
    1. Mom CH, Verweij J, Oldenhuis CN, Gietema JA, Fox NL, Miceli R, et al. Mapatumumab, a fully human agonistic monoclonal antibody that targets TRAIL-R1, in combination with gemcitabine and cisplatin: a phase I study. Clin Cancer Res. 2009;15:5584–90. doi: 10.1158/1078-0432.CCR-09-0996.
    1. Leong S, Cohen RB, Gustafson DL, Langer CJ, Camidge DR, Padavic K, et al. Mapatumumab, an antibody targeting TRAIL-R1, in combination with paclitaxel and carboplatin in patients with advanced solid malignancies: results of a phase I and pharmacokinetic study. J Clin Oncol. 2009;27:4413–21. doi: 10.1200/JCO.2008.21.7422.
    1. Plummer R, Attard G, Pacey S, Li L, Razak A, Perrett R, et al. Phase 1 and pharmacokinetic study of lexatumumab in patients with advanced cancers. Clin Cancer Res. 2007;13:6187–94. doi: 10.1158/1078-0432.CCR-07-0950.
    1. Tolcher AW, Mita M, Meropol NJ, von Mehren M, Patnaik A, Padavic K, et al. Phase I pharmacokinetic and biologic correlative study of Mapatumumab, a fully human monoclonal antibody with agonist activity to tumor necrosis factor related apoptosis-inducing ligand receptor-1. J Clin Oncol. 2007;25:1390–6. doi: 10.1200/JCO.2006.08.8898.
    1. Abdulghani J, El-Deiry WS. TRAIL receptor signaling and therapeutics. Expert Opin Ther Targets. 2010;14:1091–108. doi: 10.1517/14728222.2010.519701.
    1. Bremer E, Helfrich W. A Better TRAIL Variant for Tumor Cell-Specific Targeting? - Letter. Mol Cancer Ther. 2010;9:2853. doi: 10.1158/1535-7163.MCT-10-0621.
    1. Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 1999;5:157–63. doi: 10.1038/5517.
    1. Ganten TM, Koschny R, Sykora J, Schulze-Bergkamen H, Büchler P, Haas TL, et al. Preclinical differentiation between apparently safe and potentially hepatotoxic applications of TRAIL either alone or in combination with chemotherapeutic drugs. Clin Cancer Res. 2006;12:2640–6. doi: 10.1158/1078-0432.CCR-05-2635.
    1. Allen JE, Ferrini R, Dicker DT, Batzer G, Chen E, Oltean DI, et al. Targeting TRAIL death receptor 4 with trivalent DR4 Atrimer complexes. Mol Cancer Ther. 2012;11:2087–95. doi: 10.1158/1535-7163.MCT-12-0366.
    1. Menon LG, Kelly K, Yang HW, Kim SK, Black PM, Carroll RS, et al. Human bone marrow-derived mesenchymal stromal cells expressing S-TRAIL as a cellular delivery vehicle for human glioma therapy. Stem Cells. 2009;27:2320–30. doi: 10.1002/stem.136.
    1. Wang G, Wang X, Yu H, Wei S, Williams N, Holmes DL, et al. Small-molecule activation of the TRAIL receptor DR5 in human cancer cells. Nat Chem Biol. 2013;9:84–9. doi: 10.1038/nchembio.1153.
    1. Allen JE, El-Deiry WS. Regulation of the human TRAIL gene. Canc Biol Ther. 2012;13:1143–51. doi: 10.4161/cbt.21354.
    1. Kuribayashi K, Krigsfeld G, Wang W, Xu J, Mayes PA, Dicker DT, et al. TNFSF10 (TRAIL), a p53 target gene that mediates p53-dependent cell death. Canc Biol Ther. 2008;7:2034–8. doi: 10.4161/cbt.7.12.7460.
    1. Ghaffari S, Jagani Z, Kitidis C, Lodish HF, Khosravi-Far R. Cytokines and BCR-ABL mediate suppression of TRAIL-induced apoptosis through inhibition of forkhead FOXO3a transcription factor. Proc Natl Acad Sci. 2003;100:6523–8. doi: 10.1073/pnas.0731871100.
    1. Bunz F, Hwang PM, Torrance C, Waldman T, Zhang Y, Dillehay L. Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J Clin Investig. 1999;104:263–9. doi: 10.1172/JCI6863.
    1. Burns TF, El-Deiry WS. Identification of Inhibitors of TRAIL-induced Death (ITIDs) in the TRAIL-sensitive colon carcinoma cell line SW480 using a genetic approach. J Biol Chem. 2001;276:37879–86.
    1. Allen JE, Krigsfeld G, Mayes PA, Patel L, Dicker DT, Patel AS, et al. Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction and potent anti-tumor effects. Sci Transl Med. 2013;5:171ra17. doi: 10.1126/scitranslmed.3004828.
    1. Allen JE, El-Deiry WS. Oxaliplatin uses JNK to restore TRAIL sensitivity in cancer cells through Bcl-xL inactivation. Gastroenterology. 2011;141:430–4. doi: 10.1053/j.gastro.2011.06.026.
    1. Wu GS, Burns TF, McDonald ER, 3rd, Jiang W, Meng R, Krantz ID, et al. KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet. 2014;17:141–3. doi: 10.1038/ng1097-141.
    1. Shao R-G, Shimizu T, Pommier Y, Brefeldin A. Is a potent inducer of apoptosis in human cancer cells independently of p53. Exp Cell Res. 2014;2:190–6.
    1. Ishizawa J, Kojima K, Chachad D, Ruvolo PP, Ruvolo VR, Jacamo R, et al. ONC201 induces p53-independent apoptosis and cell cycle arrest in hematological malignancies and leukemic stem/progenitor cells by inducing ER stress and mTOR inhibition. Blood. 2014;124:3122.
    1. Allen JE, Prabhu VV, Talekar M, van den Heuvel A, Lim B, Dicker DT, et al. Genetic and pharmacological screens converge in identifying FLIP, BCL2 and IAP proteins as key regulators of sensitivity to the TRAIL-inducing anti-cancer agent ONC201/TIC10. Cancer Res. 2015. [Epub ahead of print].
    1. Wagner J, Kline CL, Pottorf RS, Nallaganchu BR, Olson GL, Dicker DT, et al. The angular structure of ONC201, a TRAIL pathway-inducing compound, determines its potent anti-cancer activity. Oncotarget. 2014;5:12728–37.
    1. Prabhu VV, Talekar M, van den Heuvel A, Lim B, Dicker DT, et al. Small molecule ONC201/TIC10 targets chemotherapy-resistant colorectal cancer stem-like cells in an Akt/Foxo3a/TRAIL-dependent manner. Cancer Res. 2015. [Epub ahead of print].

Source: PubMed

3
購読する