Skip to main content
Top
Published in: BioDrugs 5/2019

Open Access 01-10-2019 | Review Article

Mission Possible: Advances in MYC Therapeutic Targeting in Cancer

Authors: Brittany L. Allen-Petersen, Rosalie C. Sears

Published in: BioDrugs | Issue 5/2019

Login to get access

Abstract

MYC is a master transcriptional regulator that controls almost all cellular processes. Over the last several decades, researchers have strived to define the context-dependent transcriptional gene programs that are controlled by MYC, as well as the mechanisms that regulate MYC function, in an effort to better understand the contribution of this oncoprotein to cancer progression. There are a wealth of data indicating that deregulation of MYC activity occurs in a large number of cancers and significantly contributes to disease progression, metastatic potential, and therapeutic resistance. Although the therapeutic targeting of MYC in cancer is highly desirable, there remain substantial structural and functional challenges that have impeded direct MYC-targeted drug development and efficacy. While efforts to drug the ‘undruggable’ may seem futile given these challenges and considering the broad reach of MYC, significant strides have been made to identify points of regulation that can be exploited for therapeutic purposes. These include targeting the deregulation of MYC transcription in cancer through small-molecule inhibitors that induce epigenetic silencing or that regulate the G-quadruplex structures within the MYC promoter. Alternatively, compounds that disrupt the DNA-binding activities of MYC have been the long-standing focus of many research groups, since this method would prevent downstream MYC oncogenic activities regardless of upstream alterations. Finally, proteins involved in the post-translational regulation of MYC have been identified as important surrogate targets to reduce MYC activity downstream of aberrant cell stimulatory signals. Given the complex regulation of the MYC signaling pathway, a combination of these approaches may provide the most durable response, but this has yet to be shown. Here, we provide a comprehensive overview of the different therapeutic strategies being employed to target oncogenic MYC function, with a focus on post-translational mechanisms.
Literature
1.
go back to reference Kantarjian H, et al. Improved survival in chronic myeloid leukemia since the introduction of imatinib therapy: a single-institution historical experience. Blood. 2012;119(9):1981–7.PubMedPubMedCentral Kantarjian H, et al. Improved survival in chronic myeloid leukemia since the introduction of imatinib therapy: a single-institution historical experience. Blood. 2012;119(9):1981–7.PubMedPubMedCentral
2.
go back to reference Mendes D, et al. The benefit of HER2-targeted therapies on overall survival of patients with metastatic HER2-positive breast cancer–a systematic review. Breast Cancer Res. 2015;17:140.PubMedPubMedCentral Mendes D, et al. The benefit of HER2-targeted therapies on overall survival of patients with metastatic HER2-positive breast cancer–a systematic review. Breast Cancer Res. 2015;17:140.PubMedPubMedCentral
3.
go back to reference Bailey MH, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173(2):371–385.e318.PubMedPubMedCentral Bailey MH, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173(2):371–385.e318.PubMedPubMedCentral
5.
7.
go back to reference Schaub FX, et al. Pan-cancer alterations of the MYC oncogene and its proximal network across the cancer genome atlas. Cell Syst. 2018;6(3):282–300.e282.PubMedPubMedCentral Schaub FX, et al. Pan-cancer alterations of the MYC oncogene and its proximal network across the cancer genome atlas. Cell Syst. 2018;6(3):282–300.e282.PubMedPubMedCentral
8.
go back to reference Conacci-Sorrell M, McFerrin L, Eisenman RN. An overview of MYC and its interactome. Cold Spring Harb Perspect Med. 2014;4(1):a014357.PubMedPubMedCentral Conacci-Sorrell M, McFerrin L, Eisenman RN. An overview of MYC and its interactome. Cold Spring Harb Perspect Med. 2014;4(1):a014357.PubMedPubMedCentral
11.
go back to reference Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer. 2008;8(12):976–90.PubMed Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer. 2008;8(12):976–90.PubMed
14.
go back to reference Bisso A, Sabo A, Amati B. MYC in germinal center-derived lymphomas: mechanisms and therapeutic opportunities. Immunol Rev. 2019;288(1):178–97.PubMed Bisso A, Sabo A, Amati B. MYC in germinal center-derived lymphomas: mechanisms and therapeutic opportunities. Immunol Rev. 2019;288(1):178–97.PubMed
16.
go back to reference Hann SR, Eisenman RN. Proteins encoded by the human c-myc oncogene: differential expression in neoplastic cells. Mol Cell Biol. 1984;4(11):2486–97.PubMedPubMedCentral Hann SR, Eisenman RN. Proteins encoded by the human c-myc oncogene: differential expression in neoplastic cells. Mol Cell Biol. 1984;4(11):2486–97.PubMedPubMedCentral
17.
go back to reference Blackwood EM, Eisenman RN. Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. Science. 1991;251(4998):1211–7.PubMed Blackwood EM, Eisenman RN. Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. Science. 1991;251(4998):1211–7.PubMed
18.
go back to reference Blackwood EM, Luscher B, Kretzner L, Eisenman RN. The Myc: Max protein complex and cell growth regulation. Cold Spring Harb Symp Quant Biol. 1991;56:109–17.PubMed Blackwood EM, Luscher B, Kretzner L, Eisenman RN. The Myc: Max protein complex and cell growth regulation. Cold Spring Harb Symp Quant Biol. 1991;56:109–17.PubMed
19.
go back to reference Helander S, et al. Pre-anchoring of Pin1 to unphosphorylated c-Myc in a fuzzy complex regulates c-Myc activity. Structure. 2015;23(12):2267–79.PubMedPubMedCentral Helander S, et al. Pre-anchoring of Pin1 to unphosphorylated c-Myc in a fuzzy complex regulates c-Myc activity. Structure. 2015;23(12):2267–79.PubMedPubMedCentral
20.
go back to reference Kato GJ, Barrett J, Villa-Garcia M, Dang CV. An amino-terminal c-myc domain required for neoplastic transformation activates transcription. Mol Cell Biol. 1990;10(11):5914–20.PubMedPubMedCentral Kato GJ, Barrett J, Villa-Garcia M, Dang CV. An amino-terminal c-myc domain required for neoplastic transformation activates transcription. Mol Cell Biol. 1990;10(11):5914–20.PubMedPubMedCentral
21.
go back to reference Prendergast GC, Ziff EB. Methylation-sensitive sequence-specific DNA binding by the c-Myc basic region. Science. 1991;251(4990):186–9.PubMed Prendergast GC, Ziff EB. Methylation-sensitive sequence-specific DNA binding by the c-Myc basic region. Science. 1991;251(4990):186–9.PubMed
22.
go back to reference Cowling VH, Cole MD. Mechanism of transcriptional activation by the Myc oncoproteins. Semin Cancer Biol. 2006;16(4):242–52.PubMed Cowling VH, Cole MD. Mechanism of transcriptional activation by the Myc oncoproteins. Semin Cancer Biol. 2006;16(4):242–52.PubMed
23.
go back to reference Flinn EM, Busch CM, Wright AP. myc boxes, which are conserved in myc family proteins, are signals for protein degradation via the proteasome. Mol Cell Biol. 1998;18(10):5961–9.PubMedPubMedCentral Flinn EM, Busch CM, Wright AP. myc boxes, which are conserved in myc family proteins, are signals for protein degradation via the proteasome. Mol Cell Biol. 1998;18(10):5961–9.PubMedPubMedCentral
24.
go back to reference Gargano B, Amente S, Majello B, Lania L. P-TEFb is a crucial co-factor for Myc transactivation. Cell Cycle. 2007;6(16):2031–7.PubMed Gargano B, Amente S, Majello B, Lania L. P-TEFb is a crucial co-factor for Myc transactivation. Cell Cycle. 2007;6(16):2031–7.PubMed
26.
go back to reference Yada M, et al. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. EMBO J. 2004;3(10):2116–25. Yada M, et al. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. EMBO J. 2004;3(10):2116–25.
27.
go back to reference Kurland JF, Tansey WP. Myc-mediated transcriptional repression by recruitment of histone deacetylase. Cancer Res. 2008;68(10):3624–9.PubMed Kurland JF, Tansey WP. Myc-mediated transcriptional repression by recruitment of histone deacetylase. Cancer Res. 2008;68(10):3624–9.PubMed
28.
go back to reference Herbst A, et al. A conserved element in Myc that negatively regulates its proapoptotic activity. EMBO Rep. 2005;6(2):177–83.PubMedPubMedCentral Herbst A, et al. A conserved element in Myc that negatively regulates its proapoptotic activity. EMBO Rep. 2005;6(2):177–83.PubMedPubMedCentral
29.
go back to reference Gregory MA, Hann SR. c-Myc proteolysis by the ubiquitin-proteasome pathway: stabilization of c-Myc in Burkitt’s lymphoma cells. Mol Cell Biol. 2000;20(7):2423–35.PubMedPubMedCentral Gregory MA, Hann SR. c-Myc proteolysis by the ubiquitin-proteasome pathway: stabilization of c-Myc in Burkitt’s lymphoma cells. Mol Cell Biol. 2000;20(7):2423–35.PubMedPubMedCentral
30.
go back to reference Cowling VH, Chandriani S, Whitfield ML, Cole MD. A conserved Myc protein domain, MBIV, regulates DNA binding, apoptosis, transformation, and G2 arrest. Mol Cell Biol. 2006;26(11):4226–39.PubMedPubMedCentral Cowling VH, Chandriani S, Whitfield ML, Cole MD. A conserved Myc protein domain, MBIV, regulates DNA binding, apoptosis, transformation, and G2 arrest. Mol Cell Biol. 2006;26(11):4226–39.PubMedPubMedCentral
31.
go back to reference Grandori C, Cowley SM, James LP, Eisenman RN. The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol. 2000;16:653–99.PubMed Grandori C, Cowley SM, James LP, Eisenman RN. The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol. 2000;16:653–99.PubMed
32.
go back to reference Lutterbach B, Hann SR. Hierarchical phosphorylation at N-terminal transformation-sensitive sites in c-Myc protein is regulated by mitogens and in mitosis. Mol Cell Biol. 1994;14(8):5510–22.PubMedPubMedCentral Lutterbach B, Hann SR. Hierarchical phosphorylation at N-terminal transformation-sensitive sites in c-Myc protein is regulated by mitogens and in mitosis. Mol Cell Biol. 1994;14(8):5510–22.PubMedPubMedCentral
33.
go back to reference Sears R, et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 2000;14(19):2501–14.PubMedPubMedCentral Sears R, et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 2000;14(19):2501–14.PubMedPubMedCentral
34.
go back to reference Sears RC. The life cycle of C-myc: from synthesis to degradation. Cell Cycle. 2004;3(9):1133–7.PubMed Sears RC. The life cycle of C-myc: from synthesis to degradation. Cell Cycle. 2004;3(9):1133–7.PubMed
35.
go back to reference Hydbring P, et al. Phosphorylation by Cdk2 is required for Myc to repress Ras-induced senescence in cotransformation. Proc Natl Acad Sci USA. 2010;107(1):58–63.PubMed Hydbring P, et al. Phosphorylation by Cdk2 is required for Myc to repress Ras-induced senescence in cotransformation. Proc Natl Acad Sci USA. 2010;107(1):58–63.PubMed
36.
go back to reference Pulverer BJ, et al. Site-specific modulation of c-Myc cotransformation by residues phosphorylated in vivo. Oncogene. 1994;9(1):59–70.PubMed Pulverer BJ, et al. Site-specific modulation of c-Myc cotransformation by residues phosphorylated in vivo. Oncogene. 1994;9(1):59–70.PubMed
37.
go back to reference Yeh E, et al. A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat Cell Biol. 2004;6(4):308–18.PubMed Yeh E, et al. A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat Cell Biol. 2004;6(4):308–18.PubMed
38.
go back to reference Welcker M, et al. The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc Natl Acad Sci USA. 2004;101(24):9085–90.PubMedPubMedCentral Welcker M, et al. The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc Natl Acad Sci USA. 2004;101(24):9085–90.PubMedPubMedCentral
39.
go back to reference Arnold HK, et al. The Axin1 scaffold protein promotes formation of a degradation complex for c-Myc. EMBO J. 2009;28(5):500–12.PubMedPubMedCentral Arnold HK, et al. The Axin1 scaffold protein promotes formation of a degradation complex for c-Myc. EMBO J. 2009;28(5):500–12.PubMedPubMedCentral
40.
go back to reference Arnold HK, Sears RC. Protein phosphatase 2A regulatory subunit B56alpha associates with c-myc and negatively regulates c-myc accumulation. Mol Cell Biol. 2006;26(7):2832–44.PubMedPubMedCentral Arnold HK, Sears RC. Protein phosphatase 2A regulatory subunit B56alpha associates with c-myc and negatively regulates c-myc accumulation. Mol Cell Biol. 2006;26(7):2832–44.PubMedPubMedCentral
42.
go back to reference Sabo A, et al. Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature. 2014;511(7510):488–92.PubMedPubMedCentral Sabo A, et al. Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature. 2014;511(7510):488–92.PubMedPubMedCentral
43.
go back to reference Morton JP, Sansom OJ. MYC-y mice: from tumour initiation to therapeutic targeting of endogenous MYC. Mol Oncol. 2013;7(2):248–58.PubMedPubMedCentral Morton JP, Sansom OJ. MYC-y mice: from tumour initiation to therapeutic targeting of endogenous MYC. Mol Oncol. 2013;7(2):248–58.PubMedPubMedCentral
44.
go back to reference Kalkat M, et al. MYC deregulation in primary human cancers. Genes (Basel). 2017;8(6):E151. Kalkat M, et al. MYC deregulation in primary human cancers. Genes (Basel). 2017;8(6):E151.
45.
go back to reference Malempati S, et al. Aberrant stabilization of c-Myc protein in some lymphoblastic leukemias. Leukemia. 2006;20(9):1572–81.PubMedPubMedCentral Malempati S, et al. Aberrant stabilization of c-Myc protein in some lymphoblastic leukemias. Leukemia. 2006;20(9):1572–81.PubMedPubMedCentral
46.
go back to reference Zhang X, et al. Mechanistic insight into Myc stabilization in breast cancer involving aberrant Axin1 expression. Proc Natl Acad Sci USA. 2012;109(8):2790–5.PubMed Zhang X, et al. Mechanistic insight into Myc stabilization in breast cancer involving aberrant Axin1 expression. Proc Natl Acad Sci USA. 2012;109(8):2790–5.PubMed
47.
go back to reference Farrell AS, et al. Targeting inhibitors of the tumor suppressor PP2A for the treatment of pancreatic cancer. Mol Cancer Res. 2014;12(6):924–39.PubMedPubMedCentral Farrell AS, et al. Targeting inhibitors of the tumor suppressor PP2A for the treatment of pancreatic cancer. Mol Cancer Res. 2014;12(6):924–39.PubMedPubMedCentral
48.
go back to reference Hemann MT, et al. Evasion of the p53 tumour surveillance network by tumour-derived MYC mutants. Nature. 2005;436(7052):807–11.PubMedPubMedCentral Hemann MT, et al. Evasion of the p53 tumour surveillance network by tumour-derived MYC mutants. Nature. 2005;436(7052):807–11.PubMedPubMedCentral
49.
go back to reference Janghorban M, et al. Targeting c-MYC by antagonizing PP2A inhibitors in breast cancer. Proc Natl Acad Sci USA. 2014;111(25):9157–62.PubMedPubMedCentral Janghorban M, et al. Targeting c-MYC by antagonizing PP2A inhibitors in breast cancer. Proc Natl Acad Sci USA. 2014;111(25):9157–62.PubMedPubMedCentral
50.
go back to reference Schleger C, Verbeke C, Hildenbrand R, Zentgraf H, Bleyl U. c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Mod Pathol. 2002;15(4):462–9.PubMed Schleger C, Verbeke C, Hildenbrand R, Zentgraf H, Bleyl U. c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Mod Pathol. 2002;15(4):462–9.PubMed
51.
go back to reference Wang X, et al. Phosphorylation regulates c-Myc’s oncogenic activity in the mammary gland. Cancer Res. 2011;71(3):925–36.PubMedPubMedCentral Wang X, et al. Phosphorylation regulates c-Myc’s oncogenic activity in the mammary gland. Cancer Res. 2011;71(3):925–36.PubMedPubMedCentral
52.
go back to reference Farrell AS, et al. MYC regulates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma associated with poor outcome and chemoresistance. Nat Commun. 2017;8(1):1728.PubMedPubMedCentral Farrell AS, et al. MYC regulates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma associated with poor outcome and chemoresistance. Nat Commun. 2017;8(1):1728.PubMedPubMedCentral
53.
go back to reference Walz S, et al. Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature. 2014;511(7510):483–7.PubMedPubMedCentral Walz S, et al. Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature. 2014;511(7510):483–7.PubMedPubMedCentral
54.
go back to reference Soucek L, et al. Inhibition of Myc family proteins eradicates KRas-driven lung cancer in mice. Genes Dev. 2013;27(5):504–13.PubMedPubMedCentral Soucek L, et al. Inhibition of Myc family proteins eradicates KRas-driven lung cancer in mice. Genes Dev. 2013;27(5):504–13.PubMedPubMedCentral
55.
go back to reference Sansom OJ, et al. Myc deletion rescues Apc deficiency in the small intestine. Nature. 2007;446(7136):676–9.PubMed Sansom OJ, et al. Myc deletion rescues Apc deficiency in the small intestine. Nature. 2007;446(7136):676–9.PubMed
56.
go back to reference Annibali D, et al. Myc inhibition is effective against glioma and reveals a role for Myc in proficient mitosis. Nat Commun. 2014;5:4632.PubMed Annibali D, et al. Myc inhibition is effective against glioma and reveals a role for Myc in proficient mitosis. Nat Commun. 2014;5:4632.PubMed
57.
go back to reference Soucek L, Nasi S, Evan GI. Omomyc expression in skin prevents Myc-induced papillomatosis. Cell Death Differ. 2004;11(9):1038–45.PubMed Soucek L, Nasi S, Evan GI. Omomyc expression in skin prevents Myc-induced papillomatosis. Cell Death Differ. 2004;11(9):1038–45.PubMed
59.
go back to reference Jung LA, et al. OmoMYC blunts promoter invasion by oncogenic MYC to inhibit gene expression characteristic of MYC-dependent tumors. Oncogene. 2017;36(14):1911–24.PubMed Jung LA, et al. OmoMYC blunts promoter invasion by oncogenic MYC to inhibit gene expression characteristic of MYC-dependent tumors. Oncogene. 2017;36(14):1911–24.PubMed
60.
go back to reference Liu J, et al. Intrinsic disorder in transcription factors. Biochemistry. 2006;45(22):6873–88.PubMed Liu J, et al. Intrinsic disorder in transcription factors. Biochemistry. 2006;45(22):6873–88.PubMed
61.
go back to reference Tsafou K, Tiwari PB, Forman-Kay JD, Metallo SJ, Toretsky JA. targeting intrinsically disordered transcription factors: changing the paradigm. J Mol Biol. 2018;430(16):2321–41.PubMed Tsafou K, Tiwari PB, Forman-Kay JD, Metallo SJ, Toretsky JA. targeting intrinsically disordered transcription factors: changing the paradigm. J Mol Biol. 2018;430(16):2321–41.PubMed
62.
go back to reference Gao J, Xu D. Correlation between posttranslational modification and intrinsic disorder in protein. Pac Symp Biocomput. 2012; 94-103. Gao J, Xu D. Correlation between posttranslational modification and intrinsic disorder in protein. Pac Symp Biocomput. 2012; 94-103.
63.
go back to reference Poole CJ, van Riggelen J. MYC-master regulator of the cancer epigenome and transcriptome. Genes (Basel). 2017;8(5):E142. Poole CJ, van Riggelen J. MYC-master regulator of the cancer epigenome and transcriptome. Genes (Basel). 2017;8(5):E142.
64.
go back to reference Yang Z, He N, Zhou Q. Brd4 recruits P-TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol Cell Biol. 2008;28(3):967–76.PubMed Yang Z, He N, Zhou Q. Brd4 recruits P-TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol Cell Biol. 2008;28(3):967–76.PubMed
65.
66.
go back to reference Bandopadhayay P, et al. BET bromodomain inhibition of MYC-amplified medulloblastoma. Clin Cancer Res. 2014;20(4):912–25.PubMed Bandopadhayay P, et al. BET bromodomain inhibition of MYC-amplified medulloblastoma. Clin Cancer Res. 2014;20(4):912–25.PubMed
67.
go back to reference Kato F, et al. MYCL is a target of a BET bromodomain inhibitor, JQ1, on growth suppression efficacy in small cell lung cancer cells. Oncotarget. 2016;7(47):77378–88.PubMedPubMedCentral Kato F, et al. MYCL is a target of a BET bromodomain inhibitor, JQ1, on growth suppression efficacy in small cell lung cancer cells. Oncotarget. 2016;7(47):77378–88.PubMedPubMedCentral
68.
go back to reference Bhadury J, et al. BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc Natl Acad Sci USA. 2014;111(26):E2721–30.PubMedPubMedCentral Bhadury J, et al. BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc Natl Acad Sci USA. 2014;111(26):E2721–30.PubMedPubMedCentral
69.
go back to reference da Motta LL, et al. The BET inhibitor JQ1 selectively impairs tumour response to hypoxia and downregulates CA9 and angiogenesis in triple negative breast cancer. Oncogene. 2017;36(1):122–32.PubMed da Motta LL, et al. The BET inhibitor JQ1 selectively impairs tumour response to hypoxia and downregulates CA9 and angiogenesis in triple negative breast cancer. Oncogene. 2017;36(1):122–32.PubMed
70.
go back to reference Kurimchak AM, et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 2016;16(5):1273–86.PubMedPubMedCentral Kurimchak AM, et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 2016;16(5):1273–86.PubMedPubMedCentral
71.
go back to reference Leal AS, et al. Bromodomain inhibitors, JQ1 and I-BET 762, as potential therapies for pancreatic cancer. Cancer Lett. 2017;394:76–87.PubMed Leal AS, et al. Bromodomain inhibitors, JQ1 and I-BET 762, as potential therapies for pancreatic cancer. Cancer Lett. 2017;394:76–87.PubMed
72.
go back to reference Qiu H, et al. JQ1 suppresses tumor growth through downregulating LDHA in ovarian cancer. Oncotarget. 2015;6(9):6915–30.PubMedPubMedCentral Qiu H, et al. JQ1 suppresses tumor growth through downregulating LDHA in ovarian cancer. Oncotarget. 2015;6(9):6915–30.PubMedPubMedCentral
73.
go back to reference Wang J, et al. The BET bromodomain inhibitor JQ1 radiosensitizes non-small cell lung cancer cells by upregulating p21. Cancer Lett. 2017;391:141–51.PubMed Wang J, et al. The BET bromodomain inhibitor JQ1 radiosensitizes non-small cell lung cancer cells by upregulating p21. Cancer Lett. 2017;391:141–51.PubMed
74.
go back to reference Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic approach. Ann Oncol. 2017;28(8):1776–87.PubMed Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic approach. Ann Oncol. 2017;28(8):1776–87.PubMed
75.
go back to reference Kharenko OA, Hansen HC. Novel approaches to targeting BRD4. Drug Discov Today Technol. 2017;24:19–24.PubMed Kharenko OA, Hansen HC. Novel approaches to targeting BRD4. Drug Discov Today Technol. 2017;24:19–24.PubMed
76.
go back to reference Gerlach D, et al. The novel BET bromodomain inhibitor BI 894999 represses super-enhancer-associated transcription and synergizes with CDK9 inhibition in AML. Oncogene. 2018;37(20):2687–701.PubMedPubMedCentral Gerlach D, et al. The novel BET bromodomain inhibitor BI 894999 represses super-enhancer-associated transcription and synergizes with CDK9 inhibition in AML. Oncogene. 2018;37(20):2687–701.PubMedPubMedCentral
77.
go back to reference Andrews FH, et al. Dual-activity PI3 K-BRD4 inhibitor for the orthogonal inhibition of MYC to block tumor growth and metastasis. Proc Natl Acad Sci USA. 2017;114(7):E1072–80.PubMedPubMedCentral Andrews FH, et al. Dual-activity PI3 K-BRD4 inhibitor for the orthogonal inhibition of MYC to block tumor growth and metastasis. Proc Natl Acad Sci USA. 2017;114(7):E1072–80.PubMedPubMedCentral
78.
go back to reference Sun K, et al. Dual HDAC and PI3 K Inhibitor CUDC-907 downregulates MYC and suppresses growth of MYC-dependent cancers. Mol Cancer Ther. 2017;16(2):285–99.PubMed Sun K, et al. Dual HDAC and PI3 K Inhibitor CUDC-907 downregulates MYC and suppresses growth of MYC-dependent cancers. Mol Cancer Ther. 2017;16(2):285–99.PubMed
79.
go back to reference Hurley LH, Von Hoff DD, Siddiqui-Jain A, Yang D. Drug targeting of the c-MYC promoter to repress gene expression via a G-quadruplex silencer element. Semin Oncol. 2006;33(4):498–512.PubMed Hurley LH, Von Hoff DD, Siddiqui-Jain A, Yang D. Drug targeting of the c-MYC promoter to repress gene expression via a G-quadruplex silencer element. Semin Oncol. 2006;33(4):498–512.PubMed
80.
go back to reference Palumbo SL, Ebbinghaus SW, Hurley LH. Formation of a unique end-to-end stacked pair of G-quadruplexes in the hTERT core promoter with implications for inhibition of telomerase by G-quadruplex-interactive ligands. J Am Chem Soc. 2009;131(31):10878–91.PubMedPubMedCentral Palumbo SL, Ebbinghaus SW, Hurley LH. Formation of a unique end-to-end stacked pair of G-quadruplexes in the hTERT core promoter with implications for inhibition of telomerase by G-quadruplex-interactive ligands. J Am Chem Soc. 2009;131(31):10878–91.PubMedPubMedCentral
81.
go back to reference Qin Y, Rezler EM, Gokhale V, Sun D, Hurley LH. Characterization of the G-quadruplexes in the duplex nuclease hypersensitive element of the PDGF-A promoter and modulation of PDGF-A promoter activity by TMPyP4. Nucleic Acids Res. 2007;35(22):7698–713.PubMedPubMedCentral Qin Y, Rezler EM, Gokhale V, Sun D, Hurley LH. Characterization of the G-quadruplexes in the duplex nuclease hypersensitive element of the PDGF-A promoter and modulation of PDGF-A promoter activity by TMPyP4. Nucleic Acids Res. 2007;35(22):7698–713.PubMedPubMedCentral
82.
go back to reference Brown RV, Danford FL, Gokhale V, Hurley LH, Brooks TA. Demonstration that drug-targeted down-regulation of MYC in non-Hodgkins lymphoma is directly mediated through the promoter G-quadruplex. J Biol Chem. 2011;286(47):41018–27.PubMedPubMedCentral Brown RV, Danford FL, Gokhale V, Hurley LH, Brooks TA. Demonstration that drug-targeted down-regulation of MYC in non-Hodgkins lymphoma is directly mediated through the promoter G-quadruplex. J Biol Chem. 2011;286(47):41018–27.PubMedPubMedCentral
83.
go back to reference Calabrese DR, et al. Chemical and structural studies provide a mechanistic basis for recognition of the MYC G-quadruplex. Nat Commun. 2018;9(1):4229.PubMedPubMedCentral Calabrese DR, et al. Chemical and structural studies provide a mechanistic basis for recognition of the MYC G-quadruplex. Nat Commun. 2018;9(1):4229.PubMedPubMedCentral
84.
go back to reference Das T, Panda D, Saha P, Dash J. Small molecule driven stabilization of promoter G-quadruplexes and transcriptional regulation of c-MYC. Bioconjug Chem. 2018;29(8):2636–45.PubMed Das T, Panda D, Saha P, Dash J. Small molecule driven stabilization of promoter G-quadruplexes and transcriptional regulation of c-MYC. Bioconjug Chem. 2018;29(8):2636–45.PubMed
85.
go back to reference Gonzalez V, Hurley LH. The C-terminus of nucleolin promotes the formation of the c-MYC G-quadruplex and inhibits c-MYC promoter activity. Biochemistry. 2010;49(45):9706–14.PubMed Gonzalez V, Hurley LH. The C-terminus of nucleolin promotes the formation of the c-MYC G-quadruplex and inhibits c-MYC promoter activity. Biochemistry. 2010;49(45):9706–14.PubMed
86.
go back to reference Mathad RI, Hatzakis E, Dai J, Yang D. c-MYC promoter G-quadruplex formed at the 5′-end of NHE III1 element: insights into biological relevance and parallel-stranded G-quadruplex stability. Nucleic Acids Res. 2011;39(20):9023–33.PubMedPubMedCentral Mathad RI, Hatzakis E, Dai J, Yang D. c-MYC promoter G-quadruplex formed at the 5′-end of NHE III1 element: insights into biological relevance and parallel-stranded G-quadruplex stability. Nucleic Acids Res. 2011;39(20):9023–33.PubMedPubMedCentral
87.
go back to reference Hu MH, et al. Discovery of a new four-leaf clover-like ligand as a potent c-MYC transcription inhibitor specifically targeting the promoter G-quadruplex. J Med Chem. 2018;61(6):2447–59.PubMed Hu MH, et al. Discovery of a new four-leaf clover-like ligand as a potent c-MYC transcription inhibitor specifically targeting the promoter G-quadruplex. J Med Chem. 2018;61(6):2447–59.PubMed
88.
go back to reference Bouvard C, et al. Small molecule selectively suppresses MYC transcription in cancer cells. Proc Natl Acad Sci USA. 2017;114(13):3497–502.PubMedPubMedCentral Bouvard C, et al. Small molecule selectively suppresses MYC transcription in cancer cells. Proc Natl Acad Sci USA. 2017;114(13):3497–502.PubMedPubMedCentral
89.
go back to reference Zhang S, et al. Real-time monitoring of DNA G-quadruplexes in living cells with a small-molecule fluorescent probe. Nucleic Acids Res. 2018;46(15):7522–32.PubMedPubMedCentral Zhang S, et al. Real-time monitoring of DNA G-quadruplexes in living cells with a small-molecule fluorescent probe. Nucleic Acids Res. 2018;46(15):7522–32.PubMedPubMedCentral
90.
go back to reference Sutherland C, Cui Y, Mao H, Hurley LH. A mechanosensor mechanism controls the G-quadruplex/i-motif molecular switch in the MYC promoter NHE III1. J Am Chem Soc. 2016;138(42):14138–51.PubMed Sutherland C, Cui Y, Mao H, Hurley LH. A mechanosensor mechanism controls the G-quadruplex/i-motif molecular switch in the MYC promoter NHE III1. J Am Chem Soc. 2016;138(42):14138–51.PubMed
92.
go back to reference Yap JL, et al. Pharmacophore identification of c-Myc inhibitor 10074-G5. Bioorg Med Chem Lett. 2013;23(1):370–4.PubMed Yap JL, et al. Pharmacophore identification of c-Myc inhibitor 10074-G5. Bioorg Med Chem Lett. 2013;23(1):370–4.PubMed
93.
go back to reference Huang MJ, Cheng YC, Liu CR, Lin S, Liu HE. A small-molecule c-Myc inhibitor, 10058-F4, induces cell-cycle arrest, apoptosis, and myeloid differentiation of human acute myeloid leukemia. Exp Hematol. 2006;34(11):1480–9.PubMed Huang MJ, Cheng YC, Liu CR, Lin S, Liu HE. A small-molecule c-Myc inhibitor, 10058-F4, induces cell-cycle arrest, apoptosis, and myeloid differentiation of human acute myeloid leukemia. Exp Hematol. 2006;34(11):1480–9.PubMed
94.
go back to reference Berg T, et al. Small-molecule antagonists of Myc/Max dimerization inhibit Myc-induced transformation of chicken embryo fibroblasts. Proc Natl Acad Sci USA. 2002;99(6):3830–5.PubMedPubMedCentral Berg T, et al. Small-molecule antagonists of Myc/Max dimerization inhibit Myc-induced transformation of chicken embryo fibroblasts. Proc Natl Acad Sci USA. 2002;99(6):3830–5.PubMedPubMedCentral
95.
go back to reference Carabet LA, Rennie PS, Cherkasov A. Therapeutic inhibition of Myc in cancer. Structural bases and computer-aided drug discovery approaches. Int J Mol Sci. 2018;20(1):E120.PubMed Carabet LA, Rennie PS, Cherkasov A. Therapeutic inhibition of Myc in cancer. Structural bases and computer-aided drug discovery approaches. Int J Mol Sci. 2018;20(1):E120.PubMed
96.
go back to reference Castell A, et al. A selective high affinity MYC-binding compound inhibits MYC:MAX interaction and MYC-dependent tumor cell proliferation. Sci Rep. 2018;8(1):10064.PubMedPubMedCentral Castell A, et al. A selective high affinity MYC-binding compound inhibits MYC:MAX interaction and MYC-dependent tumor cell proliferation. Sci Rep. 2018;8(1):10064.PubMedPubMedCentral
97.
go back to reference Steiger D, Furrer M, Schwinkendorf D, Gallant P. Max-independent functions of Myc in Drosophila melanogaster. Nat Genet. 2008;40(9):1084–91.PubMed Steiger D, Furrer M, Schwinkendorf D, Gallant P. Max-independent functions of Myc in Drosophila melanogaster. Nat Genet. 2008;40(9):1084–91.PubMed
98.
go back to reference Struntz NB, et al. Stabilization of the Max homodimer with a small molecule attenuates Myc-driven transcription. Cell Chem Biol. 2019;26(5):711–723.e14.PubMed Struntz NB, et al. Stabilization of the Max homodimer with a small molecule attenuates Myc-driven transcription. Cell Chem Biol. 2019;26(5):711–723.e14.PubMed
99.
go back to reference Soucek L, et al. Omomyc, a potential Myc dominant negative, enhances Myc-induced apoptosis. Cancer Res. 2002;62(12):3507–10.PubMed Soucek L, et al. Omomyc, a potential Myc dominant negative, enhances Myc-induced apoptosis. Cancer Res. 2002;62(12):3507–10.PubMed
100.
go back to reference Bidwell GL 3rd, et al. Thermally targeted delivery of a c-Myc inhibitory polypeptide inhibits tumor progression and extends survival in a rat glioma model. PLoS One. 2013;8(1):e55104.PubMedPubMedCentral Bidwell GL 3rd, et al. Thermally targeted delivery of a c-Myc inhibitory polypeptide inhibits tumor progression and extends survival in a rat glioma model. PLoS One. 2013;8(1):e55104.PubMedPubMedCentral
101.
go back to reference Wang E, et al. Tumor penetrating peptides inhibiting MYC as a potent targeted therapeutic strategy for triple-negative breast cancers. Oncogene. 2019;38(1):140–50.PubMed Wang E, et al. Tumor penetrating peptides inhibiting MYC as a potent targeted therapeutic strategy for triple-negative breast cancers. Oncogene. 2019;38(1):140–50.PubMed
102.
go back to reference Beaulieu ME, et al. Intrinsic cell-penetrating activity propels Omomyc from proof of concept to viable anti-MYC therapy. Sci Transl Med. 2019;11(484):eaar5012.PubMedPubMedCentral Beaulieu ME, et al. Intrinsic cell-penetrating activity propels Omomyc from proof of concept to viable anti-MYC therapy. Sci Transl Med. 2019;11(484):eaar5012.PubMedPubMedCentral
103.
go back to reference Huang CH, et al. CDK9-mediated transcription elongation is required for MYC addiction in hepatocellular carcinoma. Genes Dev. 2014;28(16):1800–14.PubMedPubMedCentral Huang CH, et al. CDK9-mediated transcription elongation is required for MYC addiction in hepatocellular carcinoma. Genes Dev. 2014;28(16):1800–14.PubMedPubMedCentral
104.
go back to reference Bolin S, et al. Combined BET bromodomain and CDK2 inhibition in MYC-driven medulloblastoma. Oncogene. 2018;37(21):2850–62.PubMedPubMedCentral Bolin S, et al. Combined BET bromodomain and CDK2 inhibition in MYC-driven medulloblastoma. Oncogene. 2018;37(21):2850–62.PubMedPubMedCentral
105.
go back to reference Jiang H, et al. Concurrent HER or PI3 K inhibition potentiates the antitumor effect of the ERK inhibitor ulixertinib in preclinical pancreatic cancer models. Mol Cancer Ther. 2018;17(10):2144–55.PubMedPubMedCentral Jiang H, et al. Concurrent HER or PI3 K inhibition potentiates the antitumor effect of the ERK inhibitor ulixertinib in preclinical pancreatic cancer models. Mol Cancer Ther. 2018;17(10):2144–55.PubMedPubMedCentral
106.
go back to reference Morris EJ, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3(7):742–50.PubMed Morris EJ, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3(7):742–50.PubMed
107.
go back to reference Hayes TK, et al. Long-term ERK inhibition in KRAS-mutant pancreatic cancer is associated with MYC degradation and senescence-like growth suppression. Cancer Cell. 2016;29(1):75–89.PubMed Hayes TK, et al. Long-term ERK inhibition in KRAS-mutant pancreatic cancer is associated with MYC degradation and senescence-like growth suppression. Cancer Cell. 2016;29(1):75–89.PubMed
108.
go back to reference Horiuchi D, et al. MYC pathway activation in triple-negative breast cancer is synthetic lethal with CDK inhibition. J Exp Med. 2012;209(4):679–96.PubMedPubMedCentral Horiuchi D, et al. MYC pathway activation in triple-negative breast cancer is synthetic lethal with CDK inhibition. J Exp Med. 2012;209(4):679–96.PubMedPubMedCentral
109.
go back to reference Garcia-Cuellar MP, et al. Efficacy of cyclin-dependent-kinase 9 inhibitors in a murine model of mixed-lineage leukemia. Leukemia. 2014;28(7):1427–35.PubMed Garcia-Cuellar MP, et al. Efficacy of cyclin-dependent-kinase 9 inhibitors in a murine model of mixed-lineage leukemia. Leukemia. 2014;28(7):1427–35.PubMed
110.
111.
go back to reference Kauko O, O’Connor CM, Kulesskiy E, et al. PP2A inhibition is a druggable MEK inhibitor resistance mechanism in KRAS-mutant lung cancer cells. Sci Transl Med. 2018;10(450):eaaq1093.PubMedPubMedCentral Kauko O, O’Connor CM, Kulesskiy E, et al. PP2A inhibition is a druggable MEK inhibitor resistance mechanism in KRAS-mutant lung cancer cells. Sci Transl Med. 2018;10(450):eaaq1093.PubMedPubMedCentral
112.
go back to reference Risom T, et al. Differentiation-state plasticity is a targetable resistance mechanism in basal-like breast cancer. Nat Commun. 2018;9(1):3815.PubMedPubMedCentral Risom T, et al. Differentiation-state plasticity is a targetable resistance mechanism in basal-like breast cancer. Nat Commun. 2018;9(1):3815.PubMedPubMedCentral
113.
go back to reference Arnold HK, Sears RC. A tumor suppressor role for PP2A-B56alpha through negative regulation of c-Myc and other key oncoproteins. Cancer Metast Rev. 2008;27(2):147–58. Arnold HK, Sears RC. A tumor suppressor role for PP2A-B56alpha through negative regulation of c-Myc and other key oncoproteins. Cancer Metast Rev. 2008;27(2):147–58.
114.
go back to reference Eichhorn PJ, Creyghton MP, Bernards R. Protein phosphatase 2A regulatory subunits and cancer. Biochim Biophys Acta. 2009;1795(1):1–15.PubMed Eichhorn PJ, Creyghton MP, Bernards R. Protein phosphatase 2A regulatory subunits and cancer. Biochim Biophys Acta. 2009;1795(1):1–15.PubMed
115.
go back to reference Kiely M, Kiely PA. PP2A: the wolf in sheep’s clothing? Cancers (Basel). 2015;7(2):648–69. Kiely M, Kiely PA. PP2A: the wolf in sheep’s clothing? Cancers (Basel). 2015;7(2):648–69.
116.
go back to reference Sangodkar J, et al. All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J. 2016;283(6):1004–24.PubMed Sangodkar J, et al. All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J. 2016;283(6):1004–24.PubMed
117.
go back to reference O’Connor CM, Perl A, Leonard D, Sangodkar J, Narla G. Therapeutic targeting of PP2A. Int J Biochem Cell Biol. 2018;96:182–93.PubMed O’Connor CM, Perl A, Leonard D, Sangodkar J, Narla G. Therapeutic targeting of PP2A. Int J Biochem Cell Biol. 2018;96:182–93.PubMed
118.
go back to reference Ruvolo PP. The broken “Off” switch in cancer signaling: PP2A as a regulator of tumorigenesis, drug resistance, and immune surveillance. BBA Clin. 2016;6:87–99.PubMedPubMedCentral Ruvolo PP. The broken “Off” switch in cancer signaling: PP2A as a regulator of tumorigenesis, drug resistance, and immune surveillance. BBA Clin. 2016;6:87–99.PubMedPubMedCentral
119.
go back to reference Pallas DC, et al. Polyoma small and middle T antigens and SV40 small t antigen form stable complexes with protein phosphatase 2A. Cell. 1990;60(1):167–76.PubMed Pallas DC, et al. Polyoma small and middle T antigens and SV40 small t antigen form stable complexes with protein phosphatase 2A. Cell. 1990;60(1):167–76.PubMed
120.
go back to reference Chen W, Arroyo JD, Timmons JC, Possemato R, Hahn WC. Cancer-associated PP2A Aalpha subunits induce functional haploinsufficiency and tumorigenicity. Cancer Res. 2005;65(18):8183–92.PubMed Chen W, Arroyo JD, Timmons JC, Possemato R, Hahn WC. Cancer-associated PP2A Aalpha subunits induce functional haploinsufficiency and tumorigenicity. Cancer Res. 2005;65(18):8183–92.PubMed
121.
go back to reference Sablina AA, Hector M, Colpaert N, Hahn WC. Identification of PP2A complexes and pathways involved in cell transformation. Cancer Res. 2010;70(24):10474–84.PubMedPubMedCentral Sablina AA, Hector M, Colpaert N, Hahn WC. Identification of PP2A complexes and pathways involved in cell transformation. Cancer Res. 2010;70(24):10474–84.PubMedPubMedCentral
122.
go back to reference Cristobal I, et al. Deregulation of SET is Associated with tumor progression and predicts adverse outcome in patients with early-stage colorectal cancer. J Clin Med. 2019;8(3):E346.PubMed Cristobal I, et al. Deregulation of SET is Associated with tumor progression and predicts adverse outcome in patients with early-stage colorectal cancer. J Clin Med. 2019;8(3):E346.PubMed
123.
go back to reference Cristobal I, et al. Overexpression of SET is a recurrent event associated with poor outcome and contributes to protein phosphatase 2A inhibition in acute myeloid leukemia. Haematologica. 2012;97(4):543–50.PubMedPubMedCentral Cristobal I, et al. Overexpression of SET is a recurrent event associated with poor outcome and contributes to protein phosphatase 2A inhibition in acute myeloid leukemia. Haematologica. 2012;97(4):543–50.PubMedPubMedCentral
124.
go back to reference Westermarck J, Hahn WC. Multiple pathways regulated by the tumor suppressor PP2A in transformation. Trends Mol Med. 2008;14(4):152–60.PubMed Westermarck J, Hahn WC. Multiple pathways regulated by the tumor suppressor PP2A in transformation. Trends Mol Med. 2008;14(4):152–60.PubMed
125.
go back to reference Liu CY, et al. Targeting SET to restore PP2A activity disrupts an oncogenic CIP2A-feedforward loop and impairs triple negative breast cancer progression. EBioMedicine. 2019;40:263–75.PubMedPubMedCentral Liu CY, et al. Targeting SET to restore PP2A activity disrupts an oncogenic CIP2A-feedforward loop and impairs triple negative breast cancer progression. EBioMedicine. 2019;40:263–75.PubMedPubMedCentral
126.
go back to reference Khanna A, Pimanda JE, Westermarck J. Cancerous inhibitor of protein phosphatase 2A, an emerging human oncoprotein and a potential cancer therapy target. Cancer Res. 2013;73(22):6548–53.PubMed Khanna A, Pimanda JE, Westermarck J. Cancerous inhibitor of protein phosphatase 2A, an emerging human oncoprotein and a potential cancer therapy target. Cancer Res. 2013;73(22):6548–53.PubMed
127.
go back to reference Li M, Makkinje A, Damuni Z. The myeloid leukemia-associated protein SET is a potent inhibitor of protein phosphatase 2A. J Biol Chem. 1996;271(19):11059–62.PubMed Li M, Makkinje A, Damuni Z. The myeloid leukemia-associated protein SET is a potent inhibitor of protein phosphatase 2A. J Biol Chem. 1996;271(19):11059–62.PubMed
128.
go back to reference Junttila MR, et al. CIP2A inhibits PP2A in human malignancies. Cell. 2007;130(1):51–62.PubMed Junttila MR, et al. CIP2A inhibits PP2A in human malignancies. Cell. 2007;130(1):51–62.PubMed
129.
go back to reference Niemela M, et al. CIP2A signature reveals the MYC dependency of CIP2A-regulated phenotypes and its clinical association with breast cancer subtypes. Oncogene. 2012;31(39):4266–78.PubMed Niemela M, et al. CIP2A signature reveals the MYC dependency of CIP2A-regulated phenotypes and its clinical association with breast cancer subtypes. Oncogene. 2012;31(39):4266–78.PubMed
130.
go back to reference Wang J, et al. Oncoprotein CIP2A is stabilized via interaction with tumor suppressor PP2A/B56. EMBO Rep. 2017;18(3):437–50.PubMedPubMedCentral Wang J, et al. Oncoprotein CIP2A is stabilized via interaction with tumor suppressor PP2A/B56. EMBO Rep. 2017;18(3):437–50.PubMedPubMedCentral
131.
132.
go back to reference Neviani P, et al. The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein. Cancer Cell. 2005;8(5):355–68.PubMed Neviani P, et al. The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein. Cancer Cell. 2005;8(5):355–68.PubMed
133.
go back to reference Agarwal A, et al. Antagonism of SET using OP449 enhances the efficacy of tyrosine kinase inhibitors and overcomes drug resistance in myeloid leukemia. Clin Cancer Res. 2014;20(8):2092–103.PubMedPubMedCentral Agarwal A, et al. Antagonism of SET using OP449 enhances the efficacy of tyrosine kinase inhibitors and overcomes drug resistance in myeloid leukemia. Clin Cancer Res. 2014;20(8):2092–103.PubMedPubMedCentral
134.
go back to reference Matsuoka Y, Nagahara Y, Ikekita M, Shinomiya T. A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br J Pharmacol. 2003;138(7):1303–12.PubMedPubMedCentral Matsuoka Y, Nagahara Y, Ikekita M, Shinomiya T. A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br J Pharmacol. 2003;138(7):1303–12.PubMedPubMedCentral
135.
go back to reference Cristobal I, et al. Potential anti-tumor effects of FTY720 associated with PP2A activation: a brief review. Curr Med Res Opin. 2016;32(6):1137–41.PubMed Cristobal I, et al. Potential anti-tumor effects of FTY720 associated with PP2A activation: a brief review. Curr Med Res Opin. 2016;32(6):1137–41.PubMed
136.
go back to reference Stessin AM, et al. FTY720/fingolimod, an oral S1PR modulator, mitigates radiation induced cognitive deficits. Neurosci Lett. 2017;658:1–5.PubMed Stessin AM, et al. FTY720/fingolimod, an oral S1PR modulator, mitigates radiation induced cognitive deficits. Neurosci Lett. 2017;658:1–5.PubMed
137.
go back to reference McCracken AN, et al. Phosphorylation of a constrained azacyclic FTY720 analog enhances anti-leukemic activity without inducing S1P receptor activation. Leukemia. 2017;31(3):669–77.PubMed McCracken AN, et al. Phosphorylation of a constrained azacyclic FTY720 analog enhances anti-leukemic activity without inducing S1P receptor activation. Leukemia. 2017;31(3):669–77.PubMed
138.
go back to reference Gutierrez A, et al. Phenothiazines induce PP2A-mediated apoptosis in T cell acute lymphoblastic leukemia. J Clin Invest. 2014;124(2):644–55.PubMedPubMedCentral Gutierrez A, et al. Phenothiazines induce PP2A-mediated apoptosis in T cell acute lymphoblastic leukemia. J Clin Invest. 2014;124(2):644–55.PubMedPubMedCentral
139.
go back to reference Sangodkar J, et al. Activation of tumor suppressor protein PP2A inhibits KRAS-driven tumor growth. J Clin Invest. 2017;127(6):2081–90.PubMedPubMedCentral Sangodkar J, et al. Activation of tumor suppressor protein PP2A inhibits KRAS-driven tumor growth. J Clin Invest. 2017;127(6):2081–90.PubMedPubMedCentral
140.
go back to reference Allen-Petersen BL, et al. Activation of PP2A and inhibition of mTOR synergistically reduce MYC signaling and decrease tumor growth in pancreatic ductal adenocarcinoma. Cancer Res. 2019;79(1):209–19.PubMed Allen-Petersen BL, et al. Activation of PP2A and inhibition of mTOR synergistically reduce MYC signaling and decrease tumor growth in pancreatic ductal adenocarcinoma. Cancer Res. 2019;79(1):209–19.PubMed
141.
go back to reference Zhang L, et al. Eya3 partners with PP2A to induce c-Myc stabilization and tumor progression. Nat Commun. 2018;9(1):1047.PubMedPubMedCentral Zhang L, et al. Eya3 partners with PP2A to induce c-Myc stabilization and tumor progression. Nat Commun. 2018;9(1):1047.PubMedPubMedCentral
142.
go back to reference El Boustani M, et al. A guide to PIN1 function and mutations across cancers. Front Pharmacol. 2018;9:1477.PubMed El Boustani M, et al. A guide to PIN1 function and mutations across cancers. Front Pharmacol. 2018;9:1477.PubMed
143.
go back to reference Zhou XZ, Lu KP. The isomerase PIN1 controls numerous cancer-driving pathways and is a unique drug target. Nat Rev Cancer. 2016;16(7):463–78.PubMed Zhou XZ, Lu KP. The isomerase PIN1 controls numerous cancer-driving pathways and is a unique drug target. Nat Rev Cancer. 2016;16(7):463–78.PubMed
144.
145.
go back to reference Leung KW, et al. Pin1 overexpression is associated with poor differentiation and survival in oral squamous cell carcinoma. Oncol Rep. 2009;21(4):1097–104.PubMed Leung KW, et al. Pin1 overexpression is associated with poor differentiation and survival in oral squamous cell carcinoma. Oncol Rep. 2009;21(4):1097–104.PubMed
146.
go back to reference He J, et al. Overexpression of Pin1 in non-small cell lung cancer (NSCLC) and its correlation with lymph node metastases. Lung Cancer. 2007;56(1):51–8.PubMed He J, et al. Overexpression of Pin1 in non-small cell lung cancer (NSCLC) and its correlation with lymph node metastases. Lung Cancer. 2007;56(1):51–8.PubMed
147.
go back to reference Wulf GM, et al. Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. EMBO J. 2001;20(13):3459–72.PubMedPubMedCentral Wulf GM, et al. Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. EMBO J. 2001;20(13):3459–72.PubMedPubMedCentral
148.
go back to reference Lam PB, et al. Prolyl isomerase Pin1 is highly expressed in Her2-positive breast cancer and regulates erbB2 protein stability. Mol Cancer. 2008;7:91.PubMedPubMedCentral Lam PB, et al. Prolyl isomerase Pin1 is highly expressed in Her2-positive breast cancer and regulates erbB2 protein stability. Mol Cancer. 2008;7:91.PubMedPubMedCentral
149.
go back to reference Farrell AS, et al. Pin1 regulates the dynamics of c-Myc DNA binding to facilitate target gene regulation and oncogenesis. Mol Cell Biol. 2013;33(15):2930–49.PubMedPubMedCentral Farrell AS, et al. Pin1 regulates the dynamics of c-Myc DNA binding to facilitate target gene regulation and oncogenesis. Mol Cell Biol. 2013;33(15):2930–49.PubMedPubMedCentral
151.
go back to reference Su Y, et al. Post-translational modification localizes MYC to the nuclear pore basket to regulate a subset of target genes involved in cellular responses to environmental signals. Genes Dev. 2018;32(21–22):1398–419.PubMedPubMedCentral Su Y, et al. Post-translational modification localizes MYC to the nuclear pore basket to regulate a subset of target genes involved in cellular responses to environmental signals. Genes Dev. 2018;32(21–22):1398–419.PubMedPubMedCentral
152.
go back to reference Lu KP, Zhou XZ. The prolyl isomerase PIN1: a pivotal new twist in phosphorylation signalling and disease. Nat Rev Mol Cell Biol. 2007;8(11):904–16.PubMed Lu KP, Zhou XZ. The prolyl isomerase PIN1: a pivotal new twist in phosphorylation signalling and disease. Nat Rev Mol Cell Biol. 2007;8(11):904–16.PubMed
153.
go back to reference Yang D, et al. A novel controlled release formulation of the Pin1 inhibitor ATRA to improve liver cancer therapy by simultaneously blocking multiple cancer pathways. J Control Release. 2018;269:405–22.PubMed Yang D, et al. A novel controlled release formulation of the Pin1 inhibitor ATRA to improve liver cancer therapy by simultaneously blocking multiple cancer pathways. J Control Release. 2018;269:405–22.PubMed
154.
go back to reference Moore JD, Potter A. Pin1 inhibitors: pitfalls, progress and cellular pharmacology. Bioorg Med Chem Lett. 2013;23(15):4283–91.PubMed Moore JD, Potter A. Pin1 inhibitors: pitfalls, progress and cellular pharmacology. Bioorg Med Chem Lett. 2013;23(15):4283–91.PubMed
155.
go back to reference Kanaoka R, et al. Pin1 inhibitor juglone exerts anti-oncogenic effects on LNCaP and DU145 cells despite the patterns of gene regulation by Pin1 differing between these cell lines. PLoS One. 2015;10(6):e0127467.PubMedPubMedCentral Kanaoka R, et al. Pin1 inhibitor juglone exerts anti-oncogenic effects on LNCaP and DU145 cells despite the patterns of gene regulation by Pin1 differing between these cell lines. PLoS One. 2015;10(6):e0127467.PubMedPubMedCentral
156.
go back to reference Ieda N, et al. An irreversible inhibitor of peptidyl-prolyl cis/trans isomerase Pin1 and evaluation of cytotoxicity. Bioorg Med Chem Lett. 2019;29(3):353–6.PubMed Ieda N, et al. An irreversible inhibitor of peptidyl-prolyl cis/trans isomerase Pin1 and evaluation of cytotoxicity. Bioorg Med Chem Lett. 2019;29(3):353–6.PubMed
157.
go back to reference Hu YG, Shen YF, Li Y. Effect of Pin1 inhibitor juglone on proliferation, migration and angiogenic ability of breast cancer cell line MCF7Adr. J Huazhong Univ Sci Technol Med Sci. 2015;35(4):531–4. Hu YG, Shen YF, Li Y. Effect of Pin1 inhibitor juglone on proliferation, migration and angiogenic ability of breast cancer cell line MCF7Adr. J Huazhong Univ Sci Technol Med Sci. 2015;35(4):531–4.
158.
go back to reference Fila C, Metz C, van der Sluijs P. Juglone inactivates cysteine-rich proteins required for progression through mitosis. J Biol Chem. 2008;283(31):21714–24.PubMed Fila C, Metz C, van der Sluijs P. Juglone inactivates cysteine-rich proteins required for progression through mitosis. J Biol Chem. 2008;283(31):21714–24.PubMed
159.
go back to reference Chao SH, Greenleaf AL, Price DH. Juglone, an inhibitor of the peptidyl-prolyl isomerase Pin1, also directly blocks transcription. Nucleic Acids Res. 2001;29(3):767–73.PubMedPubMedCentral Chao SH, Greenleaf AL, Price DH. Juglone, an inhibitor of the peptidyl-prolyl isomerase Pin1, also directly blocks transcription. Nucleic Acids Res. 2001;29(3):767–73.PubMedPubMedCentral
160.
go back to reference Campaner E, et al. A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action. Nat Commun. 2017;8:15772.PubMedPubMedCentral Campaner E, et al. A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action. Nat Commun. 2017;8:15772.PubMedPubMedCentral
161.
go back to reference Uchida T, et al. Pin1 and Par14 peptidyl prolyl isomerase inhibitors block cell proliferation. Chem Biol. 2003;10(1):15–24.PubMed Uchida T, et al. Pin1 and Par14 peptidyl prolyl isomerase inhibitors block cell proliferation. Chem Biol. 2003;10(1):15–24.PubMed
162.
go back to reference Wei S, et al. Active Pin1 is a key target of all-trans retinoic acid in acute promyelocytic leukemia and breast cancer. Nat Med. 2015;21(5):457–66.PubMedPubMedCentral Wei S, et al. Active Pin1 is a key target of all-trans retinoic acid in acute promyelocytic leukemia and breast cancer. Nat Med. 2015;21(5):457–66.PubMedPubMedCentral
163.
go back to reference Dingar D, et al. BioID identifies novel c-MYC interacting partners in cultured cells and xenograft tumors. J Proteomics. 2015;118:95–111.PubMed Dingar D, et al. BioID identifies novel c-MYC interacting partners in cultured cells and xenograft tumors. J Proteomics. 2015;118:95–111.PubMed
164.
go back to reference Kress TR, Sabo A, Amati B. MYC: connecting selective transcriptional control to global RNA production. Nat Rev Cancer. 2015;15(10):593–607.PubMed Kress TR, Sabo A, Amati B. MYC: connecting selective transcriptional control to global RNA production. Nat Rev Cancer. 2015;15(10):593–607.PubMed
165.
go back to reference Tu WB, et al. Myc and its interactors take shape. Biochim Biophys Acta. 2015;1849(5):469–83.PubMed Tu WB, et al. Myc and its interactors take shape. Biochim Biophys Acta. 2015;1849(5):469–83.PubMed
167.
go back to reference Kim SY, Herbst A, Tworkowski KA, Salghetti SE, Tansey WP. Skp2 regulates Myc protein stability and activity. Mol Cell. 2003;11(5):1177–88.PubMed Kim SY, Herbst A, Tworkowski KA, Salghetti SE, Tansey WP. Skp2 regulates Myc protein stability and activity. Mol Cell. 2003;11(5):1177–88.PubMed
168.
go back to reference Adhikary S, et al. The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell. 2005;123(3):409–21.PubMed Adhikary S, et al. The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell. 2005;123(3):409–21.PubMed
169.
go back to reference Wasylishen AR, et al. MYC activity is negatively regulated by a C-terminal lysine cluster. Oncogene. 2014;33(8):1066–72.PubMed Wasylishen AR, et al. MYC activity is negatively regulated by a C-terminal lysine cluster. Oncogene. 2014;33(8):1066–72.PubMed
170.
go back to reference Peter S, et al. Tumor cell-specific inhibition of MYC function using small molecule inhibitors of the HUWE1 ubiquitin ligase. EMBO Mol Med. 2014;6(12):1525–41.PubMedPubMedCentral Peter S, et al. Tumor cell-specific inhibition of MYC function using small molecule inhibitors of the HUWE1 ubiquitin ligase. EMBO Mol Med. 2014;6(12):1525–41.PubMedPubMedCentral
171.
go back to reference Myant KB, et al. HUWE1 is a critical colonic tumour suppressor gene that prevents MYC signalling, DNA damage accumulation and tumour initiation. EMBO Mol Med. 2017;9(2):181–97.PubMed Myant KB, et al. HUWE1 is a critical colonic tumour suppressor gene that prevents MYC signalling, DNA damage accumulation and tumour initiation. EMBO Mol Med. 2017;9(2):181–97.PubMed
172.
go back to reference Qu H, Liu H, Jin Y, Cui Z, Han G. HUWE1 upregulation has tumor suppressive effect in human prostate cancer cell lines through c-Myc. Biomed Pharmacother. 2018;106:309–15.PubMed Qu H, Liu H, Jin Y, Cui Z, Han G. HUWE1 upregulation has tumor suppressive effect in human prostate cancer cell lines through c-Myc. Biomed Pharmacother. 2018;106:309–15.PubMed
173.
go back to reference Popov N, et al. The ubiquitin-specific protease USP28 is required for MYC stability. Nat Cell Biol. 2007;9(7):765–74.PubMed Popov N, et al. The ubiquitin-specific protease USP28 is required for MYC stability. Nat Cell Biol. 2007;9(7):765–74.PubMed
174.
go back to reference Pan J, et al. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene. 2015;34(30):3957–67.PubMed Pan J, et al. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene. 2015;34(30):3957–67.PubMed
175.
go back to reference Sun XX, et al. The nucleolar ubiquitin-specific protease USP36 deubiquitinates and stabilizes c-Myc. Proc Natl Acad Sci USA. 2015;112(12):3734–9.PubMedPubMedCentral Sun XX, et al. The nucleolar ubiquitin-specific protease USP36 deubiquitinates and stabilizes c-Myc. Proc Natl Acad Sci USA. 2015;112(12):3734–9.PubMedPubMedCentral
176.
go back to reference Fang X, et al. Deubiquitinase USP13 maintains glioblastoma stem cells by antagonizing FBXL14-mediated Myc ubiquitination. J Exp Med. 2017;214(1):245–67.PubMedPubMedCentral Fang X, et al. Deubiquitinase USP13 maintains glioblastoma stem cells by antagonizing FBXL14-mediated Myc ubiquitination. J Exp Med. 2017;214(1):245–67.PubMedPubMedCentral
177.
go back to reference Kim D, et al. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J Cell Physiol. 2017;232(12):3664–76.PubMed Kim D, et al. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J Cell Physiol. 2017;232(12):3664–76.PubMed
179.
go back to reference Nicklas S, et al. A complex of the ubiquitin ligase TRIM32 and the deubiquitinase USP7 balances the level of c-Myc ubiquitination and thereby determines neural stem cell fate specification. Cell Death Differ. 2019;26(4):728–40.PubMed Nicklas S, et al. A complex of the ubiquitin ligase TRIM32 and the deubiquitinase USP7 balances the level of c-Myc ubiquitination and thereby determines neural stem cell fate specification. Cell Death Differ. 2019;26(4):728–40.PubMed
180.
181.
go back to reference Sun XX, et al. SUMO protease SENP1 deSUMOylates and stabilizes c-Myc. Proc Natl Acad Sci US A. 2018;115(43):10983–8. Sun XX, et al. SUMO protease SENP1 deSUMOylates and stabilizes c-Myc. Proc Natl Acad Sci US A. 2018;115(43):10983–8.
182.
183.
go back to reference Dauch D, et al. A MYC-aurora kinase A protein complex represents an actionable drug target in p53-altered liver cancer. Nat Med. 2016;22(7):744–53.PubMed Dauch D, et al. A MYC-aurora kinase A protein complex represents an actionable drug target in p53-altered liver cancer. Nat Med. 2016;22(7):744–53.PubMed
185.
go back to reference Gorgun G, et al. A novel aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma. Blood. 2010;115(25):5202–13.PubMedPubMedCentral Gorgun G, et al. A novel aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma. Blood. 2010;115(25):5202–13.PubMedPubMedCentral
186.
go back to reference Pollard JR, Mortimore M. Discovery and development of aurora kinase inhibitors as anticancer agents. J Med Chem. 2009;52(9):2629–51.PubMed Pollard JR, Mortimore M. Discovery and development of aurora kinase inhibitors as anticancer agents. J Med Chem. 2009;52(9):2629–51.PubMed
187.
go back to reference Brockmann M, et al. Small molecule inhibitors of aurora-A induce proteasomal degradation of N-Myc in childhood neuroblastoma. Cancer Cell. 2016;30(2):357–8.PubMed Brockmann M, et al. Small molecule inhibitors of aurora-A induce proteasomal degradation of N-Myc in childhood neuroblastoma. Cancer Cell. 2016;30(2):357–8.PubMed
188.
go back to reference Otto T, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell. 2009;15(1):67–78.PubMed Otto T, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell. 2009;15(1):67–78.PubMed
189.
go back to reference Tayyar Y, Jubair L, Fallaha S, McMillan NAJ. Critical risk-benefit assessment of the novel anti-cancer aurora a kinase inhibitor alisertib (MLN8237): a comprehensive review of the clinical data. Crit Rev Oncol Hematol. 2017;119:59–65.PubMed Tayyar Y, Jubair L, Fallaha S, McMillan NAJ. Critical risk-benefit assessment of the novel anti-cancer aurora a kinase inhibitor alisertib (MLN8237): a comprehensive review of the clinical data. Crit Rev Oncol Hematol. 2017;119:59–65.PubMed
191.
go back to reference Shapiro GI, et al. Clinically efficacy of the BET bromodomain inhibitor TEN-010 in an open-label substudy with patients with documented NUT-midline carcinoma (NMC) [abstract no. A49]. Mol Cancer Ther. 2015;14(12 Suppl 2):A49. Shapiro GI, et al. Clinically efficacy of the BET bromodomain inhibitor TEN-010 in an open-label substudy with patients with documented NUT-midline carcinoma (NMC) [abstract no. A49]. Mol Cancer Ther. 2015;14(12 Suppl 2):A49.
192.
go back to reference Shi X, et al. JQ1: a novel potential therapeutic target. Pharmazie. 2018;73(9):491–3.PubMed Shi X, et al. JQ1: a novel potential therapeutic target. Pharmazie. 2018;73(9):491–3.PubMed
193.
go back to reference Tontsch-Grunt U, et al. Synergistic activity of BET inhibitor BI 894999 with PLK inhibitor volasertib in AML in vitro and in vivo. Cancer Lett. 2018;421:112–20.PubMed Tontsch-Grunt U, et al. Synergistic activity of BET inhibitor BI 894999 with PLK inhibitor volasertib in AML in vitro and in vivo. Cancer Lett. 2018;421:112–20.PubMed
194.
195.
go back to reference Li B, Simon MC. Molecular pathways: targeting MYC-induced metabolic reprogramming and oncogenic stress in cancer. Clin Cancer Res. 2013;19(21):5835–41.PubMed Li B, Simon MC. Molecular pathways: targeting MYC-induced metabolic reprogramming and oncogenic stress in cancer. Clin Cancer Res. 2013;19(21):5835–41.PubMed
196.
go back to reference Matkar S, et al. An epigenetic pathway regulates sensitivity of breast cancer cells to HER2 inhibition via FOXO/c-Myc axis. Cancer Cell. 2015;28(4):472–85.PubMedPubMedCentral Matkar S, et al. An epigenetic pathway regulates sensitivity of breast cancer cells to HER2 inhibition via FOXO/c-Myc axis. Cancer Cell. 2015;28(4):472–85.PubMedPubMedCentral
197.
go back to reference Bradbury RH, et al. Optimization of a series of bivalent triazolopyridazine based bromodomain and extraterminal inhibitors: the discovery of (3R)-4-[2-[4-[1-(3-methoxy-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-4-piperidyl]phen oxy]ethyl]-1,3-dimethyl-piperazin-2-one (AZD5153). J Med Chem. 2016;59(17):7801–17.PubMed Bradbury RH, et al. Optimization of a series of bivalent triazolopyridazine based bromodomain and extraterminal inhibitors: the discovery of (3R)-4-[2-[4-[1-(3-methoxy-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-4-piperidyl]phen oxy]ethyl]-1,3-dimethyl-piperazin-2-one (AZD5153). J Med Chem. 2016;59(17):7801–17.PubMed
198.
go back to reference Rhyasen GW, et al. AZD5153: a novel bivalent bet bromodomain inhibitor highly active against hematologic malignancies. Mol Cancer Ther. 2016;15(11):2563–74.PubMed Rhyasen GW, et al. AZD5153: a novel bivalent bet bromodomain inhibitor highly active against hematologic malignancies. Mol Cancer Ther. 2016;15(11):2563–74.PubMed
199.
go back to reference Shen G, et al. AZD5153 inhibits prostate cancer cell growth in vitro and in vivo. Cell Physiol Biochem. 2018;50(2):798–809.PubMed Shen G, et al. AZD5153 inhibits prostate cancer cell growth in vitro and in vivo. Cell Physiol Biochem. 2018;50(2):798–809.PubMed
200.
go back to reference Takimoto-Shimomura T, et al. Dual targeting of bromodomain-containing 4 by AZD5153 and BCL2 by AZD4320 against B-cell lymphomas concomitantly overexpressing c-MYC and BCL2. Invest New Drugs. 2019;37(2):210–22.PubMed Takimoto-Shimomura T, et al. Dual targeting of bromodomain-containing 4 by AZD5153 and BCL2 by AZD4320 against B-cell lymphomas concomitantly overexpressing c-MYC and BCL2. Invest New Drugs. 2019;37(2):210–22.PubMed
201.
go back to reference Xu K, et al. AZD5153, a novel BRD4 inhibitor, suppresses human thyroid carcinoma cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2018;499(3):531–7.PubMed Xu K, et al. AZD5153, a novel BRD4 inhibitor, suppresses human thyroid carcinoma cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2018;499(3):531–7.PubMed
202.
go back to reference Baumgart SJ, Haendler B. Exploiting epigenetic alterations in prostate cancer. Int J Mol Sci. 2017;18(5):E1017.PubMed Baumgart SJ, Haendler B. Exploiting epigenetic alterations in prostate cancer. Int J Mol Sci. 2017;18(5):E1017.PubMed
203.
go back to reference Urbanucci A, et al. Androgen receptor deregulation drives bromodomain-mediated chromatin alterations in prostate cancer. Cell Rep. 2017;19(10):2045–59.PubMedPubMedCentral Urbanucci A, et al. Androgen receptor deregulation drives bromodomain-mediated chromatin alterations in prostate cancer. Cell Rep. 2017;19(10):2045–59.PubMedPubMedCentral
204.
go back to reference Berthon C, et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016;3(4):e186–95.PubMed Berthon C, et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016;3(4):e186–95.PubMed
205.
go back to reference Coude MM, et al. BET inhibitor OTX015 targets BRD2 and BRD4 and decreases c-MYC in acute leukemia cells. Oncotarget. 2015;6(19):17698–712.PubMedPubMedCentral Coude MM, et al. BET inhibitor OTX015 targets BRD2 and BRD4 and decreases c-MYC in acute leukemia cells. Oncotarget. 2015;6(19):17698–712.PubMedPubMedCentral
206.
go back to reference Riveiro ME, et al. OTX015 (MK-8628), a novel BET inhibitor, exhibits antitumor activity in non-small cell and small cell lung cancer models harboring different oncogenic mutations. Oncotarget. 2016;7(51):84675–87.PubMedPubMedCentral Riveiro ME, et al. OTX015 (MK-8628), a novel BET inhibitor, exhibits antitumor activity in non-small cell and small cell lung cancer models harboring different oncogenic mutations. Oncotarget. 2016;7(51):84675–87.PubMedPubMedCentral
207.
go back to reference Vazquez R, et al. Promising in vivo efficacy of the BET bromodomain inhibitor OTX015/MK-8628 in malignant pleural mesothelioma xenografts. Int J Cancer. 2017;140(1):197–207.PubMed Vazquez R, et al. Promising in vivo efficacy of the BET bromodomain inhibitor OTX015/MK-8628 in malignant pleural mesothelioma xenografts. Int J Cancer. 2017;140(1):197–207.PubMed
208.
go back to reference Campbell GR, et al. Induction of autophagy by PI3 K/MTOR and PI3 K/MTOR/BRD4 inhibitors suppresses HIV-1 replication. J Biol Chem. 2018;293(16):5808–20.PubMedPubMedCentral Campbell GR, et al. Induction of autophagy by PI3 K/MTOR and PI3 K/MTOR/BRD4 inhibitors suppresses HIV-1 replication. J Biol Chem. 2018;293(16):5808–20.PubMedPubMedCentral
209.
go back to reference Shen G, Jiang M, Pu J. Dual inhibition of BRD4 and PI3 K by SF2523 suppresses human prostate cancer cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2018;495(1):567–73.PubMed Shen G, Jiang M, Pu J. Dual inhibition of BRD4 and PI3 K by SF2523 suppresses human prostate cancer cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2018;495(1):567–73.PubMed
210.
go back to reference Zhu H, et al. Dual inhibition of BRD4 and PI3 K-AKT by SF2523 suppresses human renal cell carcinoma cell growth. Oncotarget. 2017;8(58):98471–81.PubMedPubMedCentral Zhu H, et al. Dual inhibition of BRD4 and PI3 K-AKT by SF2523 suppresses human renal cell carcinoma cell growth. Oncotarget. 2017;8(58):98471–81.PubMedPubMedCentral
211.
go back to reference Zhu JX, Xiao JR. SF2523 inhibits human chondrosarcoma cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2019;511(3):559–65.PubMed Zhu JX, Xiao JR. SF2523 inhibits human chondrosarcoma cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2019;511(3):559–65.PubMed
212.
go back to reference Qin AC, Li Y, Zhou LN, Xing CG, Lu XS. Dual PI3 K-BRD4 inhibitor SF1126 inhibits colorectal cancer cell growth in vitro and in vivo. Cell Physiol Biochem. 2019;52(4):758–68.PubMed Qin AC, Li Y, Zhou LN, Xing CG, Lu XS. Dual PI3 K-BRD4 inhibitor SF1126 inhibits colorectal cancer cell growth in vitro and in vivo. Cell Physiol Biochem. 2019;52(4):758–68.PubMed
213.
go back to reference Singh AR, et al. Single agent and synergistic activity of the “first-in-class” dual PI3 K/BRD4 inhibitor SF1126 with sorafenib in hepatocellular carcinoma. Mol Cancer Ther. 2016;15(11):2553–62.PubMedPubMedCentral Singh AR, et al. Single agent and synergistic activity of the “first-in-class” dual PI3 K/BRD4 inhibitor SF1126 with sorafenib in hepatocellular carcinoma. Mol Cancer Ther. 2016;15(11):2553–62.PubMedPubMedCentral
214.
go back to reference Fu XH, et al. CUDC-907 displays potent antitumor activity against human pancreatic adenocarcinoma in vitro and in vivo through inhibition of HDAC6 to downregulate c-Myc expression. Acta Pharmacol Sin. 2019;40(5):677–88.PubMed Fu XH, et al. CUDC-907 displays potent antitumor activity against human pancreatic adenocarcinoma in vitro and in vivo through inhibition of HDAC6 to downregulate c-Myc expression. Acta Pharmacol Sin. 2019;40(5):677–88.PubMed
215.
go back to reference Oki Y, et al. CUDC-907 in relapsed/refractory diffuse large B-cell lymphoma, including patients with MYC-alterations: results from an expanded phase I trial. Haematologica. 2017;102(11):1923–30.PubMedPubMedCentral Oki Y, et al. CUDC-907 in relapsed/refractory diffuse large B-cell lymphoma, including patients with MYC-alterations: results from an expanded phase I trial. Haematologica. 2017;102(11):1923–30.PubMedPubMedCentral
216.
go back to reference Musso L, et al. c-MYC G-quadruplex binding by the RNA polymerase I inhibitor BMH-21 and analogues revealed by a combined NMR and biochemical approach. Biochim Biophys Acta Gen Subj. 2018;1862(3):615–29.PubMed Musso L, et al. c-MYC G-quadruplex binding by the RNA polymerase I inhibitor BMH-21 and analogues revealed by a combined NMR and biochemical approach. Biochim Biophys Acta Gen Subj. 2018;1862(3):615–29.PubMed
217.
go back to reference Drygin D, et al. Anticancer activity of CX-3543: a direct inhibitor of rRNA biogenesis. Cancer Res. 2009;69(19):7653–61.PubMed Drygin D, et al. Anticancer activity of CX-3543: a direct inhibitor of rRNA biogenesis. Cancer Res. 2009;69(19):7653–61.PubMed
218.
go back to reference Fukazawa T, et al. Inhibition of Myc effectively targets KRAS mutation-positive lung cancer expressing high levels of Myc. Anticancer Res. 2010;30(10):4193–200.PubMed Fukazawa T, et al. Inhibition of Myc effectively targets KRAS mutation-positive lung cancer expressing high levels of Myc. Anticancer Res. 2010;30(10):4193–200.PubMed
220.
go back to reference Stellas D, et al. Therapeutic effects of an anti-Myc drug on mouse pancreatic cancer. J Natl Cancer Inst. 2014;106(12):dju320.PubMed Stellas D, et al. Therapeutic effects of an anti-Myc drug on mouse pancreatic cancer. J Natl Cancer Inst. 2014;106(12):dju320.PubMed
221.
go back to reference Bashash D, et al. Small molecule inhibitor of c-Myc 10058-F4 inhibits proliferation and induces apoptosis in acute leukemia cells, irrespective of PTEN status. Int J Biochem Cell Biol. 2019;108:7–16.PubMed Bashash D, et al. Small molecule inhibitor of c-Myc 10058-F4 inhibits proliferation and induces apoptosis in acute leukemia cells, irrespective of PTEN status. Int J Biochem Cell Biol. 2019;108:7–16.PubMed
222.
go back to reference Follis AV, Hammoudeh DI, Daab AT, Metallo SJ. Small-molecule perturbation of competing interactions between c-Myc and Max. Bioorg Med Chem Lett. 2009;19(3):807–10.PubMed Follis AV, Hammoudeh DI, Daab AT, Metallo SJ. Small-molecule perturbation of competing interactions between c-Myc and Max. Bioorg Med Chem Lett. 2009;19(3):807–10.PubMed
223.
go back to reference Guo J, et al. Efficacy, pharmacokinetics, tisssue distribution, and metabolism of the Myc-Max disruptor, 10058–F4 [Z, E]-5-[4-ethylbenzylidine]-2-thioxothiazolidin-4-one, in mice. Cancer Chemother Pharmacol. 2009;63(4):615–25.PubMed Guo J, et al. Efficacy, pharmacokinetics, tisssue distribution, and metabolism of the Myc-Max disruptor, 10058–F4 [Z, E]-5-[4-ethylbenzylidine]-2-thioxothiazolidin-4-one, in mice. Cancer Chemother Pharmacol. 2009;63(4):615–25.PubMed
224.
go back to reference Zhang M, Fan HY, Li SC. Inhibition of c-Myc by 10058-F4 induces growth arrest and chemosensitivity in pancreatic ductal adenocarcinoma. Biomed Pharmacother. 2015;73:123–8.PubMed Zhang M, Fan HY, Li SC. Inhibition of c-Myc by 10058-F4 induces growth arrest and chemosensitivity in pancreatic ductal adenocarcinoma. Biomed Pharmacother. 2015;73:123–8.PubMed
225.
go back to reference Aksoz M, et al. c-Myc inhibitor 10074-G5 induces murine and human hematopoietic stem and progenitor cell expansion and HDR modulator Rad51 expression. Curr Cancer Drug Targets. 2019;19(6):479–94.PubMed Aksoz M, et al. c-Myc inhibitor 10074-G5 induces murine and human hematopoietic stem and progenitor cell expansion and HDR modulator Rad51 expression. Curr Cancer Drug Targets. 2019;19(6):479–94.PubMed
226.
go back to reference Wang H, et al. Disruption of Myc-Max heterodimerization with improved cell-penetrating analogs of the small molecule 10074-G5. Oncotarget. 2013;4(6):936–47.PubMedPubMedCentral Wang H, et al. Disruption of Myc-Max heterodimerization with improved cell-penetrating analogs of the small molecule 10074-G5. Oncotarget. 2013;4(6):936–47.PubMedPubMedCentral
227.
go back to reference Follis AV, Hammoudeh DI, Wang H, Prochownik EV, Metallo SJ. Structural rationale for the coupled binding and unfolding of the c-Myc oncoprotein by small molecules. Chem Biol. 2008;15(11):1149–55.PubMed Follis AV, Hammoudeh DI, Wang H, Prochownik EV, Metallo SJ. Structural rationale for the coupled binding and unfolding of the c-Myc oncoprotein by small molecules. Chem Biol. 2008;15(11):1149–55.PubMed
228.
go back to reference Hart JR, et al. Inhibitor of MYC identified in a Krohnke pyridine library. Proc Natl Acad Sci USA. 2014;111(34):12556–61.PubMedPubMedCentral Hart JR, et al. Inhibitor of MYC identified in a Krohnke pyridine library. Proc Natl Acad Sci USA. 2014;111(34):12556–61.PubMedPubMedCentral
229.
go back to reference Jeong KC, et al. Intravesical instillation of c-MYC inhibitor KSI-3716 suppresses orthotopic bladder tumor growth. J Urol. 2014;191(2):510–8.PubMed Jeong KC, et al. Intravesical instillation of c-MYC inhibitor KSI-3716 suppresses orthotopic bladder tumor growth. J Urol. 2014;191(2):510–8.PubMed
230.
go back to reference Seo HK, et al. Antitumor activity of the c-Myc inhibitor KSI-3716 in gemcitabine-resistant bladder cancer. Oncotarget. 2014;5(2):326–37.PubMedPubMedCentral Seo HK, et al. Antitumor activity of the c-Myc inhibitor KSI-3716 in gemcitabine-resistant bladder cancer. Oncotarget. 2014;5(2):326–37.PubMedPubMedCentral
231.
go back to reference Christensen DJ, et al. SET oncoprotein overexpression in B-cell chronic lymphocytic leukemia and non-Hodgkin lymphoma: a predictor of aggressive disease and a new treatment target. Blood. 2011;118(15):4150–8.PubMedPubMedCentral Christensen DJ, et al. SET oncoprotein overexpression in B-cell chronic lymphocytic leukemia and non-Hodgkin lymphoma: a predictor of aggressive disease and a new treatment target. Blood. 2011;118(15):4150–8.PubMedPubMedCentral
232.
go back to reference Enjoji S, et al. The therapeutic effects of SET/I2PP2A inhibitors on canine melanoma. J Vet Med Sci. 2015;77(11):1451–6.PubMedPubMedCentral Enjoji S, et al. The therapeutic effects of SET/I2PP2A inhibitors on canine melanoma. J Vet Med Sci. 2015;77(11):1451–6.PubMedPubMedCentral
233.
go back to reference Hu X, et al. Inhibition of Pten deficient castration resistant prostate cancer by targeting of the SET-PP2A signaling axis. Sci Rep. 2015;5:15182.PubMedPubMedCentral Hu X, et al. Inhibition of Pten deficient castration resistant prostate cancer by targeting of the SET-PP2A signaling axis. Sci Rep. 2015;5:15182.PubMedPubMedCentral
234.
go back to reference Shlomai G, et al. OP449 inhibits breast cancer growth without adverse metabolic effects. Endocr Relat Cancer. 2017;24(10):519–29.PubMedPubMedCentral Shlomai G, et al. OP449 inhibits breast cancer growth without adverse metabolic effects. Endocr Relat Cancer. 2017;24(10):519–29.PubMedPubMedCentral
235.
go back to reference Omar HA, et al. Antitumor effects of OSU-2S, a nonimmunosuppressive analogue of FTY720, in hepatocellular carcinoma. Hepatology. 2011;53(6):1943–58.PubMed Omar HA, et al. Antitumor effects of OSU-2S, a nonimmunosuppressive analogue of FTY720, in hepatocellular carcinoma. Hepatology. 2011;53(6):1943–58.PubMed
236.
go back to reference Tibaldi E, et al. Targeted activation of the SHP-1/PP2A signaling axis elicits apoptosis of chronic lymphocytic leukemia cells. Haematologica. 2017;102(8):1401–12.PubMedPubMedCentral Tibaldi E, et al. Targeted activation of the SHP-1/PP2A signaling axis elicits apoptosis of chronic lymphocytic leukemia cells. Haematologica. 2017;102(8):1401–12.PubMedPubMedCentral
237.
go back to reference Leu WJ, et al. Non-immunosuppressive triazole-based small molecule induces anticancer activity against human hormone-refractory prostate cancers: the role in inhibition of PI3 K/AKT/mTOR and c-Myc signaling pathways. Oncotarget. 2016;7(47):76995–7009.PubMedPubMedCentral Leu WJ, et al. Non-immunosuppressive triazole-based small molecule induces anticancer activity against human hormone-refractory prostate cancers: the role in inhibition of PI3 K/AKT/mTOR and c-Myc signaling pathways. Oncotarget. 2016;7(47):76995–7009.PubMedPubMedCentral
238.
go back to reference Wang S, et al. Discovery of a small molecule targeting SET-PP2A interaction to overcome BCR-ABLT315I mutation of chronic myeloid leukemia. Oncotarget. 2015;6(14):12128–40.PubMedPubMedCentral Wang S, et al. Discovery of a small molecule targeting SET-PP2A interaction to overcome BCR-ABLT315I mutation of chronic myeloid leukemia. Oncotarget. 2015;6(14):12128–40.PubMedPubMedCentral
239.
go back to reference Wu J, et al. Celastrol inhibits chondrosarcoma proliferation, migration and invasion through suppression CIP2A/c-MYC signaling pathway. J Pharmacol Sci. 2017;134(1):22–8.PubMed Wu J, et al. Celastrol inhibits chondrosarcoma proliferation, migration and invasion through suppression CIP2A/c-MYC signaling pathway. J Pharmacol Sci. 2017;134(1):22–8.PubMed
240.
go back to reference Wang H, et al. Direct inhibition of c-Myc-Max heterodimers by celastrol and celastrol-inspired triterpenoids. Oncotarget. 2015;6(32):32380–95.PubMedPubMedCentral Wang H, et al. Direct inhibition of c-Myc-Max heterodimers by celastrol and celastrol-inspired triterpenoids. Oncotarget. 2015;6(32):32380–95.PubMedPubMedCentral
241.
go back to reference Liu Z, et al. Cancerous inhibitor of PP2A is targeted by natural compound celastrol for degradation in non-small-cell lung cancer. Carcinogenesis. 2014;35(4):905–14.PubMed Liu Z, et al. Cancerous inhibitor of PP2A is targeted by natural compound celastrol for degradation in non-small-cell lung cancer. Carcinogenesis. 2014;35(4):905–14.PubMed
242.
go back to reference Chao TT, et al. TD-19, an erlotinib derivative, induces epidermal growth factor receptor wild-type nonsmall-cell lung cancer apoptosis through CIP2A-mediated pathway. J Pharmacol Exp Ther. 2014;351(2):352–8.PubMed Chao TT, et al. TD-19, an erlotinib derivative, induces epidermal growth factor receptor wild-type nonsmall-cell lung cancer apoptosis through CIP2A-mediated pathway. J Pharmacol Exp Ther. 2014;351(2):352–8.PubMed
243.
go back to reference Yu HC, et al. Erlotinib derivative inhibits hepatocellular carcinoma by targeting CIP2A to reactivate protein phosphatase 2A. Cell Death Dis. 2014;5:e1359.PubMedPubMedCentral Yu HC, et al. Erlotinib derivative inhibits hepatocellular carcinoma by targeting CIP2A to reactivate protein phosphatase 2A. Cell Death Dis. 2014;5:e1359.PubMedPubMedCentral
244.
go back to reference Liu CY, et al. EGFR-independent Elk1/CIP2A signalling mediates apoptotic effect of an erlotinib derivative TD52 in triple-negative breast cancer cells. Eur J Cancer. 2017;72:112–23.PubMed Liu CY, et al. EGFR-independent Elk1/CIP2A signalling mediates apoptotic effect of an erlotinib derivative TD52 in triple-negative breast cancer cells. Eur J Cancer. 2017;72:112–23.PubMed
245.
go back to reference Tseng LM, et al. CIP2A is a target of bortezomib in human triple negative breast cancer cells. Breast Cancer Res. 2012;14(2):R68.PubMedPubMedCentral Tseng LM, et al. CIP2A is a target of bortezomib in human triple negative breast cancer cells. Breast Cancer Res. 2012;14(2):R68.PubMedPubMedCentral
246.
go back to reference Hennig L, et al. Selective inactivation of parvulin-like peptidyl-prolyl cis/trans isomerases by juglone. Biochemistry. 1998;37(17):5953–60.PubMed Hennig L, et al. Selective inactivation of parvulin-like peptidyl-prolyl cis/trans isomerases by juglone. Biochemistry. 1998;37(17):5953–60.PubMed
247.
go back to reference D’Artista L, et al. Pin1 is required for sustained B cell proliferation upon oncogenic activation of Myc. Oncotarget. 2016;7(16):21786–98.PubMedPubMedCentral D’Artista L, et al. Pin1 is required for sustained B cell proliferation upon oncogenic activation of Myc. Oncotarget. 2016;7(16):21786–98.PubMedPubMedCentral
248.
go back to reference Hu X, et al. Prolyl isomerase PIN1 regulates the stability, transcriptional activity and oncogenic potential of BRD4. Oncogene. 2017;36(36):5177–88.PubMedPubMedCentral Hu X, et al. Prolyl isomerase PIN1 regulates the stability, transcriptional activity and oncogenic potential of BRD4. Oncogene. 2017;36(36):5177–88.PubMedPubMedCentral
249.
go back to reference Gianni M, et al. Inhibition of the peptidyl-prolyl-isomerase Pin1 enhances the responses of acute myeloid leukemia cells to retinoic acid via stabilization of RARalpha and PML-RARalpha. Cancer Res. 2009;69(3):1016–26.PubMed Gianni M, et al. Inhibition of the peptidyl-prolyl-isomerase Pin1 enhances the responses of acute myeloid leukemia cells to retinoic acid via stabilization of RARalpha and PML-RARalpha. Cancer Res. 2009;69(3):1016–26.PubMed
250.
go back to reference Wu J, et al. Momordin Ic, a new natural SENP1 inhibitor, inhibits prostate cancer cell proliferation. Oncotarget. 2016;7(37):58995–9005.PubMedPubMedCentral Wu J, et al. Momordin Ic, a new natural SENP1 inhibitor, inhibits prostate cancer cell proliferation. Oncotarget. 2016;7(37):58995–9005.PubMedPubMedCentral
251.
go back to reference Huang W, et al. Triptolide inhibits the proliferation of prostate cancer cells and down-regulates SUMO-specific protease 1 expression. PLoS One. 2012;7(5):e37693.PubMedPubMedCentral Huang W, et al. Triptolide inhibits the proliferation of prostate cancer cells and down-regulates SUMO-specific protease 1 expression. PLoS One. 2012;7(5):e37693.PubMedPubMedCentral
252.
go back to reference Macarulla T, et al. Phase I study of the selective Aurora A kinase inhibitor MLN8054 in patients with advanced solid tumors: safety, pharmacokinetics, and pharmacodynamics. Mol Cancer Ther. 2010;9(10):2844–52.PubMed Macarulla T, et al. Phase I study of the selective Aurora A kinase inhibitor MLN8054 in patients with advanced solid tumors: safety, pharmacokinetics, and pharmacodynamics. Mol Cancer Ther. 2010;9(10):2844–52.PubMed
Metadata
Title
Mission Possible: Advances in MYC Therapeutic Targeting in Cancer
Authors
Brittany L. Allen-Petersen
Rosalie C. Sears
Publication date
01-10-2019
Publisher
Springer International Publishing
Published in
BioDrugs / Issue 5/2019
Print ISSN: 1173-8804
Electronic ISSN: 1179-190X
DOI
https://doi.org/10.1007/s40259-019-00370-5

Other articles of this Issue 5/2019

BioDrugs 5/2019 Go to the issue