We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Thiazole in the targeted anticancer drug discovery

    Adileh Ayati

    Drug Design & Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran

    ,
    Saeed Emami

    *Author for correspondence:

    E-mail Address: sdemami12@gmail.com

    Department of Medicinal Chemistry & Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran

    ,
    Setareh Moghimi

    Drug Design & Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran

    &
    Alireza Foroumadi

    **Author for correspondence:

    E-mail Address: aforoumadi@yahoo.com

    Drug Design & Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran

    Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

    Published Online:https://doi.org/10.4155/fmc-2018-0416

    Cancer is known as one of the main causes of death in the world; and many compounds have been synthesized to date with potential use in cancer therapy. Thiazole is a versatile heterocycle, found in the structure of many drugs in use as well as anticancer agents. This review provides an overview of recent advances in thiazole-bearing compounds as anticancer agents with particular emphasis on their mechanism of action in cancerous cells. Chemical designs, structure–activity relationships and relevant preclinical properties have been comprehensively described.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Lagergren P, Schandl A, Aaronson NK et al. Cancer survivorship: an integral part of Europe's research agenda. Mol. Oncol. 13(3), 624–635 (2018).
    • 2. Bhat M, Poojary B, Kalal BS et al. Synthesis and evaluation of thiazolidinone–pyrazole conjugates as anticancer and antimicrobial agents. Future Med. Chem. 10(9), 1017–1036 (2018).
    • 3. Beretta GL, Cassinelli G, Pennati M, Zuco V, Gatti L. Overcoming ABC transporter-mediated multidrug resistance: the dual role of tyrosine kinase inhibitors as multitargeting agents. Eur. J. Med. Chem. 142, 271–289 (2017).
    • 4. Awasthi R, Roseblade A, Hansbro PM, Rathbone MJ, Dua K, Bebawy M. Nanoparticles in cancer treatment: opportunities and obstacles. Curr. Drug Targets 19(1), 1696–1709 (2018).
    • 5. Stewart ZA, Westfall MD, Pietenpol JA. Cell-cycle dysregulation and anticancer therapy. Trends Pharmacol. Sci. 24(3), 139–145 (2003).
    • 6. Xie XX, Li H, Wang J et al. Synthesis and anticancer evaluation effects of 1-alkyl-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl) benzo[d] thiazol-2-yl) urea as anticancer agents with low toxicity. Bioorg. Med. Chem. 23(19), 6477–6485 (2015).
    • 7. Ayati A, Emami S, Asadipour A, Shafiee A, Foroumadi A. Recent applications of 1,3-thiazole core structure in the identification of new lead compounds and drug discovery. Eur. J. Med. Chem. 97, 699–718 (2015). •• Highlights the importance of thiazole core in the new lead generation.
    • 8. Djukic M, Fesatidou M, Xenikakis I et al. In vitro antioxidant activity of thiazolidinone derivatives of 1,3-thiazole and 1,3,4-thiadiazole. Chem. Biol. Interact. 286, 119–131 (2018).
    • 9. Sharma RN, Xavier FP, Vasu KK, Chaturvedi SC, Pancholi SS. Synthesis of 4-benzyl-1,3-thiazole derivatives as potential anti-inflammatory agents: an analogue-based drug design approach. J. Enzyme Inhib. Med. Chem. 24(3), 890–897 (2009).
    • 10. Bondock S, Naser T, Ammar YA. Synthesis of some new 2-(3-pyridyl)-4,5-disubstituted thiazoles as potent antimicrobial agents. Eur. J. Med. Chem. 62, 270–279 (2013).
    • 11. Lino CI, Gonçalves de Souza I, Borelli BM et al. Synthesis, molecular modeling studies and evaluation of antifungal activity of a novel series of thiazole derivatives. Eur. J. Med. Chem. 151, 248–260 (2018).
    • 12. Osman H, Yusufzai SK, Khan MS et al. New thiazolyl-coumarin hybrids: design, synthesis, characterization, x-ray crystal structure, antibacterial and antiviral evaluation. J. Mol. Struct. 1166, 147–154 (2018).
    • 13. Ahangar N, Ayati A, Alipour E, Pashapour A, Foroumadi A, Emami S. 1‐[(2‐arylthiazol‐4‐yl) methyl] azoles as a new class of anticonvulsants: design, synthesis, in vivo screening, and in silico drug‐like properties. Chem. Biol. Drug Des. 78(5), 844–852 (2011).
    • 14. Mishra CB, Kumari S, Tiwari M. Thiazole: a promising heterocycle for the development of potent CNS active agents. Eur. J. Med. Chem. 92, 1–34 (2015).
    • 15. Leoni A, Locatelli A, Morigi R, Rambaldi M. Novel thiazole derivatives: a patent review (2008–2012, Part 1). Expert Opin. Ther. Patents 24(2), 201–216 (2014). • Comprehensive review on thiazole derivatives.
    • 16. Puszkiel A, Noé G, Bellesoeur A et al. Clinical pharmacokinetics and pharmacodynamics of dabrafenib. Clin. Pharmacokinet. 58(4), 451–467 (2019).
    • 17. Keating GM. Dasatinib: a review in chronic myeloid leukaemia and Ph+ acute lymphoblastic leukaemia. Drugs 77(1), 85–96 (2017).
    • 18. Honore S, Pasquier E, Braguer D. Understanding microtubule dynamics for improved cancer therapy. Cell Mol. Life Sci. 62(24), 3039–3056 (2005). •• Highlights the importance of our knowledge about microtubule dynamics and cancer therapy.
    • 19. Bhattacharyya B, Panda D, Gupta S, Banerjee M. Anti‐mitotic activity of colchicine and the structural basis for its interaction with tubulin. Med. Res. Rev. 28(1), 155–183 (2008).
    • 20. Mirzaei H, Emami S. Recent advances of cytotoxic chalconoids targeting tubulin polymerization: synthesis and biological activity. Eur. J. Med. Chem. 121, 610–639 (2016).
    • 21. Lu Y, Chen J, Xiao M, Li W, Miller DD. An overview of tubulin inhibitors that interact with the colchicine binding site. Pharm. Res. 29(11), 2943–2971 (2012). •• Comprehensive review of tubulin polymerization inhibitors.
    • 22. Lin CM, Ho HH, Pettit GR, Hamel E. Antimitotic natural products combretastatin A-4 and combretastatin A-2: studies on the mechanism of their inhibition of the binding of colchicine to tubulin. Biochemistry 28(17), 6984–6991 (1989).
    • 23. Chaudhary A, Pandeya SN, Kumar P et al. Combretastatin A-4 analogs as anticancer agents. Mini Rev. Med. Chem. 7(12), 1186–1205 (2007).
    • 24. Mahboobi S, Sellmer A, Ho H, Schmidt M, Maier T. [4-(imidazol-1-yl)thiazol-2-yl]phenylamines. A novel class of highly potent colchicine site binding tubulin inhibitors: synthesis and cytotoxic activity on selected human cancer cell lines. J. Med. Chem. 49(19), 5769–5776 (2006).
    • 25. Romagnoli R, Baraldi PG, Carrion MD et al. 2-arylamino-4-amino-5-aroylthiazoles. “One-pot” synthesis and biological evaluation of a new class of inhibitors of tubulin polymerization. J. Med. Chem. 52(17), 5551–5555 (2009).
    • 26. Romagnoli R, Baraldi PG, Cara CL et al. One-pot synthesis and biological evaluation of 2-pyrrolidinyl-4-amino-5-(3′, 4′, 5′-trimethoxybenzoyl) thiazole: a unique, highly active antimicrotubule agent. Eur. J. Med. Chem. 46(12), 6015–6024 (2011).
    • 27. Romagnoli R, Baraldi PG, Salvador MK et al. Discovery and optimization of a series of 2-aryl-4-amino-5-(3′, 4′, 5′-trimethoxybenzoyl) thiazoles as novel anticancer agents. J. Med. Chem. 55(11), 5433–5445 (2012).
    • 28. Romagnoli R, Baraldi PG, Brancale A et al. Convergent synthesis and biological evaluation of 2-amino-4-(3′, 4′, 5′-trimethoxyphenyl)-5-aryl thiazoles as microtubule targeting agents. J. Med. Chem. 54(14), 5144–5153 (2011).
    • 29. Romagnoli R, Giovanni P, Kimatrai M et al. Synthesis and biological evaluation of 2-substituted-4-(3′, 4′, 5′-trimethoxy phenyl)-5-aryl thiazoles as anticancer agents. Bioorg. Med. Chem. 20(24), 7083–7094 (2012).
    • 30. Banimustafa M, Kheirollahi A, Safavi M et al. Synthesis and biological evaluation of 3-(trimethoxyphenyl)-2 (3H)-thiazole thiones as combretastatin analogs. Eur. J. Med. Chem. 70, 692–702 (2013).
    • 31. Sasse F, Steinmetz H, Schupp T et al. Argyrins, immunosuppressive cyclic peptides from myxobacteria. I. Production, isolation, physico-chemical and biological properties. J. Antibiot. 55(6), 543–551 (2002).
    • 32. Sani M, Lazzari P, Folini M et al. Synthesis and superpotent anticancer activity of tubulysins carrying non-hydrolysable N-substituents on tubuvaline. Chemistry 23(24), 5842–5850 (2017).
    • 33. Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy. Cell 108(2), 153–164 (2002).
    • 34. Satchell PG, Gutmann JL, Witherspoon DE. Apoptosis: an introduction for the endodontist. Int. Endod. J. 36(4), 237–245 (2003).
    • 35. Tolcher AW. Regulators of apoptosis as anticancer targets. Hematol. Oncol. Clin. North Am. 16(5), 1255–1267 (2002).
    • 36. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin. Cancer Res. 13(24), 7271–7279 (2007).
    • 37. de Bruin EC, Medema JP. Apoptosis and non-apoptotic deaths in cancer development and treatment response. Cancer Treat. Rev. 34(8), 737–749 (2008). •• Highlights the role of apoptosis in cancer development and cancer therapy.
    • 38. Tantak MP, Wang J, Prakash R, Kumar A, Shah K, Kumar D. 2-(3-indolyl)-N-arylthiazole-4-carboxamides: synthesis and evaluation of antibacterial and anticancer activities. Bioorg. Med. Chem. Lett. 25(19), 4225–4231 (2015).
    • 39. Yempala T, Sriram D, Yogeeswari P, Kantevari S. Molecular hybridization of bioactives: synthesis and antitubercular evaluation of novel dibenzofuran embodied homoisoflavonoids via Baylis–Hillman reaction. Bioorg. Med. Chem. Lett. 22(24), 7426–7430 (2012).
    • 40. Wu Z, Ding N, Tang Y, Ye J, Peng J, Hu A. Synthesis and antitumor activity of novel N-(5-benzyl-4-(tert-butyl)thiazol-2-yl)-2-(piperazin-1-yl)acetamides. Res. Chem. Intermed. 43(8), 4833–4850 (2017).
    • 41. da Santana TI, Barbosa MO, Gomes PATM, da Cruz ACN, da Silva TG, Leite ACL. Synthesis, anticancer activity and mechanism of action of new thiazole derivatives. Eur. J. Med. Chem. 144, 874–886 (2018).
    • 42. dos Santos Silva TD, Bomfim LM, da Cruz Rodrigues ACB et al. Anti-liver cancer activity in vitro and in vivo induced by 2-pyridyl 2,3-thiazole derivatives. Toxicol. Appl. Pharmacol. 329, 212–223 (2017).
    • 43. dos Santos TA, da Silva AC, Silva EB et al. Antitumor and immunomodulatory activities of thiosemicarbazones and 1,3-thiazoles in Jurkat and HT-29 cells. Biomed. Pharmacother. 82, 555–560 (2016).
    • 44. He H, Wang X, Shi L et al. Synthesis, antitumor activity and mechanism of action of novel 1,3-thiazole derivatives containing hydrazide – hydrazone and carboxamide moiety. Bioorg. Med. Chem. Lett. 26(14), 3263–3270 (2016).
    • 45. Ayati A, Esmaeili R, Moghimi S et al. Synthesis and biological evaluation of 4-amino-5-cinnamoylthiazoles as chalcone-like anticancer agents. Eur. J. Med. Chem. 145, 404–412 (2018).
    • 46. Ayati A, Oghabi Bakhshaiesh T, Moghimi S et al. Synthesis and biological evaluation of new coumarins bearing 2,4-diaminothiazole-5-carbonyl moiety. Eur. J. Med. Chem. 155, 483–491 (2018).
    • 47. Vanhaesebroeck B, Leevers SJ, Timms J et al. Synthesis and function of 3-phosphorylated inositol lipids. Annu. Rev. Biochem. 70, 535–602 (2001).
    • 48. Hayakawa M, Kaizawa H, Kawaguchi K et al. Synthesis and biological evaluation of imidazo [1,2-a] pyridine derivatives as novel PI3 kinase p110α inhibitors. Bioorg. Med. Chem. 15(1), 403–412 (2007).
    • 49. Bruce I, Akhlaq M, Bloomfield GC et al. Development of isoform selective PI3-kinase inhibitors as pharmacological tools for elucidating the PI3K pathway. Bioorg. Med. Chem. Lett. 22(17), 5445–5450 (2012). •• Described progress toward isoform selective phosphatidylinositol-3-kinase inhibitors using an automated parallel synthesis strategy.
    • 50. Furet P, Guagnano V, Fairhurst RA et al. Discovery of NVP-BYL719 a potent and selective phosphatidylinositol-3 kinase alpha inhibitor selected for clinical evaluation. Bioorg. Med. Chem. Lett. 23(13), 3741–3748 (2013).
    • 51. Fairhurst RA, Imbach-Weese P, Gerspacher M et al. Identification and optimisation of a 4′,5-bisthiazole series of selective phosphatidylinositol-3 kinase alpha inhibitors. Bioorg. Med. Chem. Lett. 25(17), 3569–3574 (2015).
    • 52. Fairhurst RA, Gerspacher M, Imbach-weese P et al. Identification and optimisation of 4,5-dihydrobenzo series of selective phosphatidylinositol-3 kinase alpha inhibitors. Bioorg. Med. Chem. Lett. 25(17), 3575–3581 (2015).
    • 53. Oka Y, Yabuuchi T, Fujii Y, Ohtake H, Wakahara S. Discovery and optimization of a series of 2-aminothiazole-oxazoles as potent phosphoinositide 3-kinase c inhibitors. Bioorg. Med. Chem. Lett. 22(24), 7534–7538 (2012).
    • 54. Oka Y, Yabuuchi T, Oi T, Kuroda S, Fujii Y, Ohtake H. (TASP0415914) as an orally potent phosphoinositide 3-kinase c inhibitor for the treatment of inflammatory diseases. Bioorg. Med. Chem. 21(24), 7578–7583 (2013).
    • 55. Certal V, Halley F, Virone-oddos A et al. Preparation and optimization of new 4-(2-(indolin-1-yl)-2-oxoethyl)-2-morpholinothiazole-5-carboxylic acid and amide derivatives as potent and selective PI3Kβ inhibitors. Bioorg. Med. Chem. Lett. 24(6), 1506–1510 (2014).
    • 56. Hundsdörfer C, Hemmerling H, Götz C et al. Indeno [1,2-b] indole derivatives as a novel class of potent human protein kinase CK2 inhibitors. Bioorg. Med. Chem. 20(7), 2282–2289 (2012).
    • 57. Pinhero R, Liaw P, Bertens K, Yankulov K. Three cyclin-dependent kinases preferentially phosphorylate different parts of the C-terminal domain of the large subunit of RNA polymerase II. Eur. J. Biochem. 271(5), 1004–1014 (2004).
    • 58. Guerra B, Issinger OG. Protein kinase CK2 in human diseases. Curr. Med. Chem. 15(19), 1870–1886 (2008). • Contains considerable information on the potential roles of CK2 in various diseases including cancer.
    • 59. Kim KS, Kimball SD, Misra RN et al. Discovery of aminothiazole inhibitors of cyclin-dependent kinase 2: synthesis, x-ray crystallographic analysis, and biological activities. J. Med. Chem. 45(18), 3905–3927 (2002).
    • 60. Misra RN, Xiao H, Kim KS et al. N-(cycloalkylamino)acyl-2-aminothiazole Inhibitors of cyclin-dependent kinase piperidinecarboxamide (BMS-387032), a highly efficacious and selective antitumor agent. J. Med. Chem. 47(7), 1719–1728 (2004).
    • 61. Choong IC, Serafimova I, Fan J et al. A diaminocyclohexyl analog of SNS-032 with improved permeability and bioavailability properties. Bioorg. Med. Chem. Lett. 18(21), 5763–5765 (2008).
    • 62. Misra RN, Xiao H, Williams DK et al. Synthesis and biological activity of N-aryl-2-aminothiazoles: potent pan inhibitors of cyclin-dependent kinases. Bioorg. Med. Chem. Lett. 14(11), 2973–2977 (2004).
    • 63. Wu SY, McNae I, Kontopidis G et al. Discovery of a novel family of CDK Inhibitors with the program LIDAEUS: structural basis for ligand-induced disordering of the activation loop. Structure 11(3), 399–410 (2003).
    • 64. Wang S, Meades C, Wood G et al. 2-anilino-4-(thiazol-5-yl) pyrimidine CDK inhibitors: synthesis, SAR analysis, x-ray crystallography, and biological activity. J. Med. Chem. 47(7), 1662–1675 (2004).
    • 65. McIntyre NA, McInnes C, Griffiths G et al. Design, synthesis, and evaluation of 2-methyl- and 2-amino- N-aryl-4,5-dihydrothiazolo-[4,5-h] quinazolin-8-amines as ring-constrained 2-anilino-4-(thiazol-5-yl) pyrimidine cyclin-dependent kinase inhibitors. J. Med. Chem. 53(4), 2136–2145 (2010).
    • 66. Shao H, Shi S, Foley DW et al. Synthesis, structure-activity relationship and biological evaluation of 2,4,5-trisubstituted pyrimidine CDK inhibitors as potential anti-tumour agents. Eur. J. Med. Chem. 70, 447–455 (2013).
    • 67. Schonbrunn E, Betzi S, Alam R et al. Development of highly potent and selective diaminothiazole inhibitors of cyclin-dependent kinases. J. Med. Chem. 56(10), 3768–3782 (2013). • Interesting article in the field of thiazole inhibitors of cyclin-dependent kinases.
    • 68. Lv PC, Zhou CF, Chen J et al. Design, synthesis and biological evaluation of thiazolidinone derivatives as potential EGFR and HER-2 kinase inhibitors. Bioorg. Med. Chem. 18(1), 314–319 (2010).
    • 69. Lichtenberger BM, Tan PK, Niederleithner H, Ferrara N, Petzelbauer P, Sibilia M. Autocrine VEGF signaling synergizes with EGFR in tumor cells to promote epithelial cancer development. Cell 140(2), 268–279 (2010).
    • 70. Qian Y, Zhang H, Zhang H, Xu C, Zhao J, Zhu H. Synthesis, molecular modeling, and biological evaluation of cinnamic acid metronidazole ester derivatives as novel anticancer agents. Bioorg. Med. Chem. 18(14), 4991–4966 (2010).
    • 71. Chandregowda V, Rao GV, Reddy GC. Convergent approach for commercial synthesis of gefitinib and erlotinib. Org. Process Res. Dev. 11(5), 813–816 (2007).
    • 72. Lv PC, Li DD, Li QS, Lu X, Xiao ZP, Zhu HL. Synthesis, molecular docking and evaluation of thiazolyl-pyrazoline derivatives as EGFR TK inhibitors and potential anticancer agents. Bioorg. Med. Chem. Lett. 21(18), 5374–5377 (2011).
    • 73. Yuan J, Wang S, Luo Z et al. Synthesis and biological evaluation of compounds which contain pyrazole, thiazole and naphthalene ring as antitumor agents. Bioorg. Med. Chem. Lett. 24(10), 2324–2328 (2014).
    • 74. El-Serwy WS, Mohamed NA, Salah W, Nossier ES, Mahmoud K. Synthesis, molecular modeling studies and biological evaluation of novel pyrazole derivatives as antitumor and EGFR inhibitors. IJPT 8(4), 25192–25209 (2016).
    • 75. Perrot-Applanat M, Di Benedetto M. Autocrine functions of VEGF in breast tumor cells: adhesion, survival, migration and invasion. Cell Adh. Migr. 6(6), 547–553 (2012). •• Contains considerable information about functions of VEGF.
    • 76. Song G, Li Y, Jiang G. Role of VEGF/VEGFR in the pathogenesis of leukemias and as treatment targets. Oncol. Rep. 28(6), 1935–1944 (2012).
    • 77. Bilodeau MT, Rodman LD, McGaughey GB et al. The discovery of N-(1,3-thiazol-2-yl) pyridin-2-amines as potent inhibitors of KDR kinase. Bioorg. Med. Chem. Lett. 14(11), 2941–2945 (2004). • Design of new lead compounds as selective kinase domain region kinase inhibitors.
    • 78. Bilodeau MT, Balitza AE, Koester TJ et al. Potent N-(1,3-thiazol-2-yl)pyridin-2-amine vascular endothelial growth factor receptor tyrosine kinase inhibitors with excellent pharmacokinetics and low affinity for the hERG ion channel. J. Med. Chem. 47(25), 6363–6372 (2004).
    • 79. Sisko JT, Tucker TJ, Bilodeau MT et al. Potent 2-[(pyrimidin-4-yl)amine}-1,3-thiazole-5-carbonitrile-based inhibitors of VEGFR-2 (KDR) kinase. Bioorg. Med. Chem. Lett. 16(5), 1146–1150 (2006).
    • 80. Kiselyov AS, Piatnitski E, Semenov VV. N-(aryl)-4-(azolylethyl)thiazole-5-carboxamides: novel potent inhibitors of VEGF receptors I and II. Bioorg. Med. Chem. Lett. 16(3), 602–606 (2006).
    • 81. Wickens P, Kluender H, Dixon J et al. SAR of a novel “anthranilamide like” series of VEGFR-2, multi protein kinase inhibitors for the treatment of cancer. Bioorg. Med. Chem. Lett. 17(15), 4378–4381 (2007).
    • 82. Abou-Seri SM, Eldehna WM, Ali MM, Abou El Ella DA. 1-piperazinylphthalazines as potential VEGFR-2 inhibitors and anticancer agents: synthesis and in vitro biological evaluation. Eur. J. Med. Chem. 107, 165–179 (2016).
    • 83. El-Miligy MM, Abd El Razik HA, Abu-Serie MM. Synthesis of piperazine-based thiazolidinones as VEGFR2 tyrosine kinase inhibitors inducing apoptosis. Future Med. Chem. 9(15), 1709–1729 (2017).
    • 84. Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ-specific colonization. Nat. Rev. Cancer. 9(4), 274–284 (2009).
    • 85. Scott RW, Hooper S, Crighton D et al. LIM kinases are required for invasive path generation by tumor and tumor-associated stromal cells. J. Cell Biol. 191(1), 169–185 (2010).
    • 86. He L, Seitz SP, Trainor GL et al. Modulation of cofilin phosphorylation by inhibition of the Lim family kinases. Bioorg. Med. Chem. Lett. 22(18), 5995–5998 (2012).
    • 87. Charles MD, Brookfield JL, Ekwuru CT et al. Discovery, development and SAR of aminothiazoles as LIMK inhibitors with cellular anti-invasive properties. J. Med. Chem. 58(20), 8309–8313 (2015). •• Interesting article about discovery of aminothiazoles as LIMK inhibitors.
    • 88. Brooks CL, Gu W. p53 ubiquitination: Mdm2 and beyond. Mol. Cell. 21(3), 307–315 (2006).
    • 89. Reverdy C, Conrath S, Lopez R, Sippl W, Harpon J. Discovery of specific inhibitors of human USP7/HAUSP deubiquitinating enzyme. Chem. Biol. 19(4), 467–477 (2012).
    • 90. Chauhan D, Tian Z, Nicholson B et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell. 22(3), 345–358 (2013).
    • 91. Altun M, Kramer HB, Willems LI et al. Activity-based chemical proteomics accelerates inhibitor development for deubiquitylating enzymes. Chem. Biol. 18(11), 1401–1412 (2011).
    • 92. Chen C, Song J, Wang J et al. Synthesis and biological evaluation of thiazole derivatives as novel USP7 inhibitors. Bioorg. Med. Chem. Lett. 27(4), 845–849 (2017).
    • 93. Gonzalez AZ, Li Z, Beck HP et al. Novel inhibitors of the MDM2-p53 interaction featuring hydrogen bond acceptors as carboxylic acid isosteres. J. Med. Chem. 57(7), 2963–2988 (2014).
    • 94. Issaeva N, Bozko P, Enge M et al. Small molecule RITA binds to p53, blocks p53–HDM-2 interaction and activates p53 function in tumors. Nat. Med. 10(12), 1321–1328 (2004).
    • 95. Sangfelt O, Selivanova G. Correction ablation of key oncogenic pathways by RITA-reactivated p53 is required for efficient apoptosis. Cancer Cell. 31(5), 724–726 (2017).
    • 96. Wanzel M, Vischedyk JB, Gittler MP et al. CRISPR-Cas9-based target validation for p53-reactivating model compounds. Nat. Chem. Biol. 12(1), 22–28 (2016).
    • 97. Pietkiewicz AL, Zhang Y, Rahimi MN, Stramandinoli M, Teusner M, McAlpine SR. RITA mimics: synthesis and mechanistic evaluation of asymmetric linked trithiazoles. ACS Med. Chem. Lett. 8(4), 401–406 (2017).
    • 98. Aliabadi A, Shamsa F, Ostad SN et al. Synthesis and biological evaluation of 2-phenylthiazole-4-carboxamide derivatives as anticancer agents. Eur. J. Med. Chem. 45, 5384–5389 (2010).
    • 99. Fallah-Tafti A, Foroumadi A, Tiwari R et al. Thiazolyl N-benzyl-substituted acetamide derivatives: synthesis, SRC kinase inhibitory and anticancer activities. Eur. J. Med. Chem. 46, 4853–4858 (2011).
    • 100. Vij N. AAA ATPase p97/VCP: cellular functions, disease and therapeutic potential role of VCP in inflammatory signalling and ER stress. J. Cell Mol. Med. 12(6), 2511–2518 (2008).
    • 101. Bursavich MG, Parker DP, Willardsen JA et al. 2-anilino-4-aryl-1,3-thiazole inhibitors of valosin-containing protein (VCP or p97). Bioorg. Med. Chem. Lett. 20(5), 1677–1679 (2010).
    • 102. Bramati A, Girelli S, Torri V et al. Efficacy of biological agents in metastatic triple-negative breast cancer. Cancer Treat. Rev. 40(5), 605–613 (2014).
    • 103. Zhou W, Huang A, Zhang Y et al. Design and optimization of hybrid of 2,4-diaminopyrimidine and arylthiazole scaffold as anticancer cell proliferation and migration agents. Eur. J. Med. Chem. 96, 269–280 (2015).