Drug Res (Stuttg) 2017; 67(03): 138-148
DOI: 10.1055/s-0042-119992
Review
© Georg Thieme Verlag KG Stuttgart · New York

Pyridazine Based Scaffolds as Privileged Structures in anti-Cancer Therapy

Maiy Youssef Jaballah
1   Faculty of Pharmacy, Pharmaceutical Chemistry Department, Ain Shams University, Cairo, Egypt
,
Rabah Taha Serya
1   Faculty of Pharmacy, Pharmaceutical Chemistry Department, Ain Shams University, Cairo, Egypt
,
Khaled Abouzid
1   Faculty of Pharmacy, Pharmaceutical Chemistry Department, Ain Shams University, Cairo, Egypt
› Author Affiliations
Further Information

Publication History

received 07 September 2016

accepted 24 October 2016

Publication Date:
10 January 2017 (online)

Abstract

Pyridazines, their oxo derivatives; pyridazinone as well as fused bi- or tricyclic pyridazine containing scaffolds are key structural features of many biologically active compounds with diverse pharmacological applications, including cancer therapy. Since protein kinases play prominent role in tumor biology, the inhibition of its signaling pathway is considered an effective therapeutic option for the treatment of cancer.

Based on the various advantages of pyridazines in drug design including modulation of the physico-chemical properties, improving ADME and toxicity profile as well as easy and diverse synthetic methods of access, makes them an invaluable tool for designing compounds as future drugs for targeted cancer treatment.

In this review, we have compiled and discussed the anticancer potential of pyridazine based scaffold, with special focus on those targeting protein kinase inhibition.

 
  • References

  • 1 Asif M. Some recent approaches of biologically active substituted pyridazine and phthalazine drugs. Curr Med Chem 2012; 19: 2984-2991
  • 2 Sotelo E et al. Pyridazines. Part XXIX: synthesis and platelet aggregation inhibition activity of 5-substituted-6-phenyl-3 (2H)-pyridazinones. Novel aspects of their biological actions. Bioorganic & medicinal chemistry 2002; 10: 2873-2882
  • 3 Coelho A, Sotelo E, Raviña E. Pyridazine derivatives. Part 33: Sonogashira approaches in the synthesis of 5-substituted-6-phenyl-3 (2H)-pyridazinones. Tetrahedron 2003; 59: 2477-2484
  • 4 Garkani-Nejad Z, Poshteh-Shirani M. Prediction of antihypertensive activity of pyridazinone derivatives through multivariate image analysis applied to QSAR. Medicinal Chemistry Research 2013; 22: 3389-3397
  • 5 Coudert P et al. Synthesis and evaluation of the aldose reductase inhibitory activity of new diaryl pyridazine-3-ones. J Pharm Belg 1991; 46: 375-380
  • 6 Allerton CM et al. Design and synthesis of pyridazinone-based 5-HT(2C) agonists. Bioorg Med Chem Lett 2009; 19: 5791-5795
  • 7 Schudt C et al. Zardaverine as a selective inhibitor of phosphodiesterase isozymes. Biochem Pharmacol 1991; 42: 153-162
  • 8 Siddiqui AA, Mishra R, Shaharyar M. Synthesis, characterization and antihypertensive activity of pyridazinone derivatives. Eur J Med Chem 2010; 45: 2283-2290
  • 9 Rubat C et al. Anticonvulsant activity of 3-oxo-5-substituted benzylidene-6-methyl-(4H)-2-pyridazinylacetamides and 2-pyridazinylacetylhydrazides. Chem Pharm Bull (Tokyo) 1990; 38: 3009-3013
  • 10 Sircar I et al. Cardiotonic agents. 7. Inhibition of separated forms of cyclic nucleotide phosphodiesterase from guinea pig cardiac muscle by 4,5-dihydro-6-[4-(1H-imidazol-1-yl)phenyl]-3(2H)-pyridazinones and related compounds. Structure-activity relationships and correlation with in vivo positive inotropic activity. Journal of Medicinal Chemistry 1987; 30: 1955-1962
  • 11 Deeb A, El-Mariah F, Hosny M. Pyridazine derivatives and related compounds. Part 13: Synthesis and antimicrobial activity of some pyridazino[3’,4’:3,4]pyrazolo[5,1-c]-1,2,4-triazines. Bioorg Med Chem Lett 2004; 14: 5013-5017
  • 12 Akahane A et al. Discovery of 6-oxo-3-(2-phenylpyrazolo[1,5–a]pyridin-3-yl)-1(6H)- pyridazinebutanoic acid (FK 838): a novel non-xanthine adenosine A1 receptor antagonist with potent diuretic activity. J Med Chem 1999; 42: 779-783
  • 13 Demirayak S et al. Some pyridazinone and phthalazinone derivatives and their vasodilator activities. Archives of Pharmacal Research 2004; 27: 13-18
  • 14 Hagiwara M et al. Effect of 1-(3-chloroanilino)-4-phenylphthalazine (MY-5445), a specific inhibitor of cyclic GMP phosphodiesterase, on human platelet aggregation. J Pharmacol Exp Ther 1984; 228: 467-471
  • 15 Murty MSR et al. Synthesis and preliminary evaluation activity studies of novel 4-(aryl/heteroaryl-2-ylmethyl)-6-phenyl-2-[3-(4-substituted-piperazine-1-yl)propyl]pyridazin-3(2H)-one derivatives as anticancer agents. Medicinal Chemistry Research 2012; 21: 3161-3169
  • 16 Malinka W. Synthesis of some pyrrolo[3,4-d]pyridazinones and their preliminary anticancer, antimycobacterial and CNS screening. Die Pharmazie 2001; 56: 384-389
  • 17 Malinka W, Redzicka A, Lozach O. New derivatives of pyrrolo[3,4-d]pyridazinone and their anticancer effects. Farmaco 2004; 59: 457-462
  • 18 Michaelis L, Granick S.. Some properties of phenanthrene semiquinone. Journal of the American Chemical Society 1948; 70: 624-627
  • 19 Wermuth CG. Are pyridazines privileged structures?. MedChemComm 2011; 2: 935-941
  • 20 Rognan D et al. Optically active benzamides as predictive tools for mapping the dopamine D2 receptor. Eur J Pharmacol 1990; 189: 59-70
  • 21 Markman M et al. Expanded experience with an intradermal skin test to predict for the presence or absence of carboplatin hypersensitivity. Journal of clinical oncology 21.24 2003; 4611-4614
  • 22 Graham L. An introduction to medicinal chemistry. 5th ed. Patrick and o.u. Press; 2009
  • 23 Pazdur R. Cancer management: a multidisciplinary approach: medical, surgical & radiation oncology, Edn. 5th ed. 2001
  • 24 Hanahan D, Weinberg RA. The Hallmarks of Cancer. Cell 2000; 100: 57-70
  • 25 Hartinger CG, Dyson PJ. Bioorganometallic chemistry-from teaching paradigms to medicinal applications. Chemical Society Reviews 2009; 38: 391-401
  • 26 Kelland L. Broadening the clinical use of platinum drug-based chemotherapy with new analogues. Expert Opinion on Investigational Drugs 2007; 16: 1009-1021
  • 27 Dyson PJ, Sava G. Metal-based antitumour drugs in the post genomic era. Dalton Transactions 2006; 1929-1933
  • 28 Todd RC, Lippard SJ. Inhibition of transcription by platinum antitumor compounds. Metallomics 2009; 1: 280-291
  • 29 Komeda S et al. New isomeric azine-bridged dinuclear platinum(ii) complexes circumvent cross-resistance to cisplatin. Journal of Medicinal Chemistry 2003; 46: 1210-1219
  • 30 Brouwers EEM et al. The application of inductively coupled plasma mass spectrometry in clinical pharmacological oncology research. Mass Spectrometry Reviews 2008; 27: 67-100
  • 31 Csókás D et al. 2,3-Dihydroimidazo[1,2–b]ferroceno[d]pyridazines and a 3,4-dihydro-2H-pyrimido[1,2–b]ferroceno[d]pyridazine: Synthesis, structure and in vitro antiproliferation activity on selected human cancer cell lines. Journal of Organometallic Chemistry 2014; 750: 41-48
  • 32 Galisteo J et al. Synthesis and cytotoxic activity of a new potential DNA bisintercalator: 1,4-Bis{3-[N-(4-chlorobenzo[g]phthalazin-1-yl)aminopropyl]}piperazine. Bioorganic & Medicinal Chemistry 2010; 18: 5301-5309
  • 33 Antoci V et al. Hybrid anticancer 1,2-diazine derivatives with multiple mechanism of action. Part 3 [4,5] Medical Hypotheses 2014; 82: 11-15
  • 34 Auerbach W, Auerbach R. Angiogenesis inhibition: a review. Pharmacol Ther 1994; 63: 265-311
  • 35 Deplanque G, Harris AL. Anti-angiogenic agents: clinical trial design and therapies in development. Eur J Cancer 2000; 36 (13 Spec No) 1713-1724
  • 36 Siemann DW. The unique characteristics of tumor vasculature and preclinical evidence for its selective disruption by Tumor-Vascular Disrupting Agents. Cancer Treat Rev 2011; 37: 63-74
  • 37 Bongartz J-P et al. Synthesis and anti-angiogenic activity of 6-(1,2,4-thiadiazol-5-yl)-3-amino pyridazine derivatives. Bioorganic & medicinal chemistry letters 2002; 12: 589-591
  • 38 Gross S et al. Targeting cancer with kinase inhibitors. The Journal of Clinical Investigation 125: 1780-1789
  • 39 Paul MK, Mukhopadhyaya AK. Tyrosine kinase – Role and significance in Cancer. International Journal of medicinal sciences 2004; 1: 101-115
  • 40 Dhanasekaran N, Premkumar Reddy E. Signaling by dual specificity kinases. Oncogene 1998; 17 (11 Reviews) 1447-1455
  • 41 Besant PG, Tan E, Attwood PV. Mammalian protein histidine kinases. The International Journal of Biochemistry & Cell Biology 2003; 35: 297-309
  • 42 Olsson A-K et al. VEGF receptor signalling? in control of vascular function. Nat Rev Mol Cell Biol 2006; 7: 359-371
  • 43 Rahimi N. Vascular endothelial growth factor receptors: Molecular mechanisms of activation and therapeutic potentials. Experimental Eye Research 2006; 83: 1005-1016
  • 44 Miyamoto N et al. Design, synthesis, and evaluation of imidazo[1,2–b]pyridazine derivatives having a benzamide unit as novel VEGFR2 kinase inhibitors. Bioorg Med Chem 2012; 20: 7051-7058
  • 45 Awazu Y et al. Anti-angiogenic and anti-tumor effects of TAK-593, a potent and selective inhibitor of vascular endothelial growth factor and platelet-derived growth factor receptor tyrosine kinase. Cancer Science 2013; 104: 486-494
  • 46 Iwata H et al. Biochemical characterization of TAK-593, a novel VEGFR/PDGFR inhibitor with a two-step slow binding mechanism. Biochemistry 2011; 50: 738-751
  • 47 Matsumoto S et al. Structure-based design, synthesis, and evaluation of imidazo[1,2–b]pyridazine and imidazo[1,2–a]pyridine derivatives as novel dual c-Met and VEGFR2 kinase inhibitors. Bioorg Med Chem 2013; 21: 7686-7698
  • 48 Huang WS et al. Discovery of 3-[2-(imidazo[1,2–b]pyridazin-3-yl)ethynyl]-4-methyl-N-{4-[(4-methylpiperazin-1-y l)methyl]-3-(trifluoromethyl)phenyl}benzamide (AP24534), a potent, orally active pan-inhibitor of breakpoint cluster region-abelson (BCR-ABL) kinase including the T315I gatekeeper mutant. J Med Chem 2010; 53: 4701-4719
  • 49 Auger KR et al. PDGF-dependent tyrosine phosphorylation stimulates production of novel polyphosphoinositides in intact cells. Cell 1989; 57: 167-175
  • 50 Cully M et al. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer 2006; 6: 184-192
  • 51 Vogt PK et al. Cancer-specific mutations in phosphatidylinositol 3-kinase. Trends Biochem Sci 2007; 32: 342-349
  • 52 Karakas B, Bachman KE, Park BH. Mutation of the PIK3CA oncogene in human cancers. Br J Cancer 2006; 94: 455-459
  • 53 Parrish et al. Discovery of GSK2126458, a highly potent inhibitor of pi3k and the mammalian target of rapamycin 2010
  • 54 Mills GB et al. Expression of TTK, a novel human protein kinase, is associated with cell proliferation. J Biol Chem 1992; 267: 16000-16006
  • 55 Lauze E et al. Yeast spindle pole body duplication gene MPS1 encodes an essential dual specificity protein kinase. Embo j 1995; 14: 1655-1663
  • 56 Naud S et al. Structure-based design of orally bioavailable 1H-pyrrolo[3,2-c]pyridine inhibitors of mitotic kinase monopolar spindle 1 (MPS1). J Med Chem 2013; 56: 10045-10065
  • 57 Kusakabe K-i et al. Discovery of imidazo[1,2–b]pyridazine derivatives: selective and orally available Mps1 (TTK) kinase inhibitors exhibiting remarkable antiproliferative activity. J Med Chem 2015; 58: 1760-1775
  • 58 Sherr CJ. Cancer cell cycles. Science 1996; 274: 1672-1677
  • 59 Sherr CJ. The Pezcoller lecture: cancer cell cycles revisited. Cancer Res 2000; 60: 3689-3695
  • 60 Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 1999; 13: 1501-1512
  • 61 Hirai H, Kawanishi N, Iwasawa Y. Recent advances in the development of selective small molecule inhibitors for cyclin-dependent kinases. Curr Top Med Chem 2005; 5: 167-179
  • 62 Brana MF et al. Pyrazolo[3,4-c]pyridazines as novel and selective inhibitors of cyclin-dependent kinases. J Med Chem 2005; 48: 6843-6854
  • 63 Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009; 324: 1029-1033
  • 64 Mazurek S et al. Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin Cancer Biol 2005; 15: 300-308
  • 65 Dombrauckas JD, Santarsiero BD, Mesecar AD. Structural basis for tumor pyruvate kinase M2 allosteric regulation and catalysis. Biochemistry 2005; 44: 9417-9429
  • 66 Jiang J-k et al. Evaluation of thieno[3,2–b]pyrrole[3,2-d]pyridazinones as activators of the tumor cell specific M2 isoform of pyruvate kinase. Bioorganic & Medicinal Chemistry Letters 2010; 20: 3387-3393
  • 67 Kumar A et al. Crystal structures of proto-oncogene kinase Pim1: a target of aberrant somatic hypermutations in diffuse large cell lymphoma. J Mol Biol 2005; 348: 183-193
  • 68 Pogacic V et al. Structural analysis identifies imidazo[1,2–b]pyridazines as PIM kinase inhibitors with in vitro antileukemic activity. Cancer Res 2007; 67: 6916-6924
  • 69 Gourley E et al. A potent small molecule PIM kinase inhibitor with activity in cell lines from hematological and solid malignancies. Cancer Research 68.9 Supplement 2008; 4974-4974
  • 70 Chen LS, Redkar WW. S et al. Pim kinase inhibitor, sgi-1776, induces apoptosis in CLL lymphocytes. in ASH Annual Meeting Abstracts 2008; 4199
  • 71 Brown VI et al. Inhibiting PIM-1 Is Effective in Vitro and in Vivo against ALL: A Novel Mechanistic and Potentially Clinically Relevant Druggable Target. Blood 112.11 2008; 1922-1922
  • 72 Grey R et al. Structure-based design of 3-aryl-6-amino-triazolo[4,3–b]pyridazine inhibitors of Pim-1 kinase. Bioorg Med Chem Letts 2009; 19: 3019-3022
  • 73 Swords R et al. The Pim kinases: new targets for drug development. Curr Drug Targets 2011; 12: 2059-2066
  • 74 Boccaccio C, Comoglio PM.. Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer 2006; 6: 637-645
  • 75 Comoglio PM, Giordano S, Trusolino L. Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov 2008; 7: 504-516
  • 76 Buchanan SG et al. SGX523 is an exquisitely selective, ATP-competitive inhibitor of the MET receptor tyrosine kinase with antitumor activity in vivo. Mol Cancer Ther 2009; 8: 3181-3190
  • 77 Park BH et al. KRC-327, a selective novel inhibitor of c-Met receptor tyrosine kinase with anticancer activity. Cancer Lett 2013; 331: 158-166
  • 78 Park BH et al. KRC-327, a selective novel inhibitor of c-Met receptor tyrosine kinase with anticancer activity. Cancer letters 331.2 2013; 158-166
  • 79 Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer 2004; 4: 937-947
  • 80 Sparano JA. Ras/Raf/MEK Inhibitors, in Molecular Targeting in Oncology. Kaufman HL, Wadler S, Antman K.. Editors. Totowa, NJ: Humana Press; 2008: 55-73
  • 81 Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature 2001; 411: 355-365
  • 82 Sakai N, SIMiyamoto N, Hirayama T. Application WP. Ed. 2008
  • 83 Park H et al. Identification of novel BRAF kinase inhibitors with structure-based virtual screening. Bioorganic & medicinal chemistry letters 21.19 2011; 5753-5756
  • 84 Park H, Jeong Y, Hong S. Structure-based de novo design and biochemical evaluation of novel BRAF kinase inhibitors. Bioorganic & Medicinal Chemistry Letters 2012; 22: 1027-1030
  • 85 Mahindroo N, Punchihewa C, Fujii N. Hedgehog-Gli Signaling Pathway Inhibitors as Anticancer Agents. Journal of Medicinal Chemistry 2009; 52: 3829-3845