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

Indole derivatives (2010–2020) as versatile tubulin inhibitors: synthesis and structure–activity relationships

    Fatima Naaz

    Department of Chemistry, School of Chemical & Lifescience, Jamia Hamdard, New Delhi, 110062, India

    ,
    Kumari Neha

    Department of Pharmaceutical Chemistry, DPSRU, New Delhi, 110017, India

    ,
    Md Rafi Haider

    Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India

    &
    Syed Shafi

    Department of Chemistry, School of Chemical & Lifescience, Jamia Hamdard, New Delhi, 110062, India

    Department of Chemistry, School of Chemical & Lifescience, Jamia Hamdard, New Delhi, 110062, India

    Published Online:https://doi.org/10.4155/fmc-2020-0385

    Tubulin inhibitors are conjugates that interfere with the dynamic equilibrium of the polymerization and depolymerization of microtubules. Among all the reported conjugates, indole moiety is one of the most significant classes for the development of new drug candidates for cancer therapy. Due to their presence in a wide range of natural as well as synthetic antitubulin agents, indole has become a versatile scaffold in research, and various synthetic and semisynthetic indole-based antitubulin agents have been identified and reported. The present article focuses on the reported indole-based tubulin inhibitors of synthetic origin from last the decade. Synthesis, structure–activity relationships and biological activities of synthetic indole derivatives along with brief updates on their antitubulin activity are presented.

    References

    • 1. Taber DF, Tirunahari PK. Indole synthesis: a review and proposed classification. Tetrahedron 67(38), 7195–7210 (2011).
    • 2. Heravi Majid M, Rohani S, Zadsirjan V, Zahedi N. Fischer indole synthesis applied to the total synthesis of natural products. RSC Adv. 7(83), 52852–52887 (2017).
    • 3. Dadashpour S, Emami S. Indole in the target-based design of anticancer agents: a versatile scaffold with diverse mechanisms. Eur. J. Med. Chem. 150, 9–29 (2018).
    • 4. Barden TC. Indoles: industrial, agricultural and over-the-counter uses. Top Heterocycl. Chem. 26, 31–46 (2010).
    • 5. Chadha N, Silakari O. Indoles as therapeutics of interest in medicinal chemistry: bird's eye view. Eur. J. Med. Chem. 134, 159–184 (2017).
    • 6. Wan Y, Li Y, Yan C, Yan M, Tang Z. Indole: a privileged scaffold for the design of anti-cancer agents. Eur. J. Med. Chem. 183, 111691 (2019).
    • 7. Han Y, Dong W, Guo Q, Li X, Huang L. The importance of indole and azaindole scaffold in the development of antitumor agents. Eur. J. Med. Chem. 203, 112506 (2020).
    • 8. Sharma S, Gupta MK, Saxena A, Singh Bedi PM. Thiazolidinone constraint combretastatin analogs as novel antitubulin agents: design, synthesis, biological evaluation and docking studies. Anti-Cancer Agents Med. Chem. 11, 230–240 (2017).
    • 9. Neeraj Mahindroo J-PL, Chang† Jang-Yang, Hsieh Hsing-Pang. Antitubulin agents for the treatment of cancer – a medicinal chemistry update. Expert Opin. Ther. Patents 16(5), 647–691 (2006).
    • 10. Singh H, Kumar M, Nepali K et al. Triazole tethered C5-curcuminoid-coumarin based molecular hybrids as novel antitubulin agents: design, synthesis, biological investigation and docking studies. Eur. J. Med. Chem. 116, 102–115 (2016).
    • 11. Nepali Kunal, Ojha R, Sharma S, Preet Bedi MS, Kanaya Dhar L. Tubulin inhibitors: a patent survey. Recent Pat. Anti-Cancer Drug Discov. 9(2), 176–220 (2014).
    • 12. Sharma S, Gupta MK, Saxena AK, Bedi PM. Triazole linked mono carbonyl curcumin-isatin bifunctional hybrids as novel anti tubulin agents: design, synthesis, biological evaluation and molecular modeling studies. Bioorg. Med. Chem. 23(22), 7165–7180 (2015).
    • 13. Mishra RC. Microtubule binding natural substances in cancer chemotherapy. Research Signpost 37(661), 269–282 (2011).
    • 14. Moudi M, Go R, Yien CYS, Nazre M. Vinca alkaloids. Int. J. Prev. Med. 4(11), 1231–1235 (2013).
    • 15. Naaz F, Haider MR, Shafi S, Yar MS. Anti-tubulin agents of natural origin: targeting taxol, vinca, and colchicine binding domains. Eur. J. Med. Chem. 171, 310–331 (2019).
    • 16. Morita H, Shimbo K, Kobayashi JI. Antimitotic activity of moroidin, a bicyclic peptide from the seeds of Celosia argentea. Bioorg. Med. Chem. Lett. 10, 469–471 (2000).
    • 17. Dou X, Dong B. Origins and bioactivities of natural compounds derived from marine ascidians and their symbionts. Mar. Drugs 17(12), 670 (2019).
    • 18. 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).
    • 19. Wienecke A, Bacher G. Indibulin, a novel microtubule inhibitor, discriminates between mature neuronal and nonneuronal tubulin. Cancer Res. 69(1), 171–177 (2009).
    • 20. Colley HE, Muthana M, Danson SJ et al. An orally bioavailable, indole-3-glyoxylamide based series of tubulin polymerization inhibitors showing tumor growth inhibition in a mouse xenograft model of head and neck cancer. J. Med. Chem. 58(23), 9309–9333 (2015).
    • 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).
    • 22. Shetty RS, Lee Y, Liu B et al. Synthesis and pharmacological evaluation of N-(3-(1H-Indol-4-yl)-5-(2-methoxyisonicotinoyl)phenyl)methanesulfonamide (LP-261), a potent antimitotic agent. J. Med. Chem. 54(1), 179–200 (2011).
    • 23. Kuo CC, Hsieh HP, Pan WY et al. BPR0L075, a novel synthetic indole compound with antimitotic activity in human cancer cells, exerts effective antitumoral activity in vivo. Cancer Res. 64(13), 4621–4628 (2004).
    • 24. Ahn S, Duke C, Herring C et al. I-387, a novel antimitotic indole, displays a potent in vitro and in vivo antitumor activity with less neurotoxicity. Mol. Cancer Ther. 9, 2859–2868 (2010).
    • 25. Hu CB, Chen CP, Yeh TK et al. BPR0C261 is a novel orally active antitumor agent with antimitotic and anti-angiogenic activities. Cancer Sci. 102(1), 182–191 (2011).
    • 26. Li WT, Yeh TK, Song JS et al. BPR0C305, an orally active microtubule-disrupting anticancer agent. Anticancer Drugs 24(10), 1047–1057 (2013).
    • 27. Lai MJ, Chang JY, Lee HY et al. Synthesis and biological evaluation of 1-(4′-Indolyl and 6′-Quinolinyl) indoles as a new class of potent anticancer agents. Eur. J. Med. Chem. 46(9), 3623–3629 (2011).
    • 28. Mullagiri K, Nayak VL, Sunkari S et al. New (3-(1H-benzo[d]imidazol-2-yl))/(3-(3H-imidazo[4,5-b]pyridin-2-yl))-(1H-indol-5-yl)(3,4,5-trimethoxyphenyl)methanone conjugates as tubulin polymerization inhibitors. MedChemComm. 9(2), 275–281 (2018).
    • 29. Hwang DJ, Wang J, Li W, Miller DD. Structural optimization of indole derivatives acting at colchicine binding site as potential anticancer agents. ACS Med. Chem. Lett. 6(9), 993–997 (2015).
    • 30. Zhai X, Wang X, Wang J et al. Discovery and optimization of novel 5-indolyl-7-arylimidazo[1,2-a]pyridine-8-carbonitrile derivatives as potent antitubulin agents targeting colchicine-binding site. Sci. Rep. 7, 43398 (2017).
    • 31. Coluccia A, Sabbadin D, Brancale A. Molecular modelling studies on arylthioindoles as potent inhibitors of tubulin polymerization. Eur. J. Med. Chem. 46(8), 3519–3525 (2011).
    • 32. La Regina G, Bai R, Rensen W et al. Design and synthesis of 2-heterocyclyl-3-arylthio-1H-indoles as potent tubulin polymerization and cell growth inhibitors with improved metabolic stability. J. Med. Chem. 54(24), 8394–8406 (2011).
    • 33. La Regina G, Bai R, Rensen WM et al. Toward highly potent cancer agents by modulating the C-2 group of the arylthioindole class of tubulin polymerization inhibitors. J. Med. Chem. 56(1), 123–149 (2013).
    • 34. La Regina G, Bai R, Coluccia A et al. New 6- and 7-heterocyclyl-1H-indole derivatives as potent tubulin assembly and cancer cell growth inhibitors. Eur. J. Med. Chem. 152, 283–297 (2018).
    • 35. Zhao DG, Chen J, Du YR et al. Synthesis and structure–activity relationships of N-methyl-5,6,7-trimethoxylindoles as novel antimitotic and vascular disrupting agents. J. Med. Chem. 56(4), 1467–1477 (2013).
    • 36. Macdonough MT, Strecker TE, Hamel E et al. Synthesis and biological evaluation of indole-based, anti-cancer agents inspired by the vascular disrupting agent 2-(3′-hydroxy-4′-methoxyphenyl)-3-(3″,4″,5″-trimethoxybenzoyl)-6-methoxyindole (OXi8006). Bioorg. Med. Chem. 21(21), 6831–6843 (2013).
    • 37. Lee HY, Lee JF, Kumar S et al. 3-Aroylindoles display antitumor activity in vitro and in vivo: effects of N1-substituents on biological activity. Eur. J. Med. Chem. 125, 1268–1278 (2017).
    • 38. Cai M, Hu J, Tian J, Yan H, Zheng C-G, Hu W-L. Novel hybrids from N-hydroxyarylamide and indole ring through click chemistry as histone deacetylase inhibitors with potent antitumor activities. Chinese Chem. Lett. 26 (2015).
    • 39. Diao PC, Li Q, Hu MJ et al. Synthesis and biological evaluation of novel indole-pyrimidine hybrids bearing morpholine and thiomorpholine moieties. Eur. J. Med. Chem. 134, 110–118 (2017).
    • 40. Chennamaneni S, Gan C, Lama R, Zhong B, Su B. Indomethacin derivatives as tubulin stabilizers to inhibit cancer cell proliferation. Bioorg. Med. Chem. 24(2), 277–285 (2016).
    • 41. Mahal K, Biersack B, Schruefer S et al. Combretastatin A-4 derived 5-(1-methyl-4-phenyl-imidazol-5-yl)indoles with superior cytotoxic and anti-vascular effects on chemoresistant cancer cells and tumors. Eur. J. Med. Chem. 118, 9–20 (2016).
    • 42. Colley HE, Muthana M, Danson SJ et al. An orally bioavailable,indole-3-glyoxylamide based series of tubulin polymerization inhibitors showing tumor growth inhibition in a mouse xenograft model of head and neck cancer. J. Med. Chem. 58(23), 9309–9333 (2015).
    • 43. Guggilapu SD, Lalita G, Reddy TS et al. Synthesis of C(5)-tethered indolyl-3-glyoxylamide derivatives as tubulin polymerization inhibitors. Eur. J. Med. Chem. 128, 1–12 (2017).
    • 44. Naaz F, Preeti Pallavi MC, Shafi S, Mulakayala N, Shahar Yar M, Sampath Kumar HM. 1,2,3-triazole tethered Indole-3-glyoxamide derivatives as multiple inhibitors of 5-LOX, COX-2 & tubulin: their anti-proliferative & anti-inflammatory activity. Bioorg. Chem. 81, 1–20 (2018).
    • 45. Kumar D, Maruthi Kumar N, Tantak MP, Ogura M, Kusaka E, Ito T. Synthesis and identification of α-cyano bis(indolyl)chalcones as novel anticancer agents. Bioorg. Med. Chem. Lett. 24(22), 5170–5174 (2014).
    • 46. Das Mukherjee D, Kumar NM, Tantak MP et al. Development of novel bis(indolyl)-hydrazide–hydrazone derivatives as potent microtubule-targeting cytotoxic agents against A549 lung cancer cells. Biochemistry 55(21), 3020–3035 (2016).
    • 47. Yan J, Chen J, Zhang S, Hu J, Huang L, Li X. Synthesis, evaluation, and mechanism study of novel indole-chalcone derivatives exerting effective antitumor activity through microtubule destabilization in vitro and in vivo. J. Med. Chem. 59(11), 5264–5283 (2016).
    • 48. Zhang S, An B, Li J et al. Synthesis and evaluation of selenium-containing indole chalcone and diarylketone derivatives as tubulin polymerization inhibition agents. Org. Biomol. Chem. 15(35), 7404–7410 (2017).
    • 49. Wang G, Li C, He L et al. Design, synthesis and biological evaluation of a series of pyrano chalcone derivatives containing indole moiety as novel anti-tubulin agents. Bioorg. Med. Chem. 22(7), 2060–2079 (2014).
    • 50. Mirzaei H, Shokrzadeh M, Modanloo M, Ziar A, Riazi GH, Emami S. New indole-based chalconoids as tubulin-targeting antiproliferative agents. Bioorg. Chem. 75, 86–98 (2017).
    • 51. Andreani A, Granaiola M, Locatelli A et al. Cytotoxic activities of substituted 3-(3,4,5-trimethoxybenzylidene)-1,3-dihydroindol-2-ones and studies on their mechanisms of action. Eur. J. Med. Chem. 64, 603–612 (2013).
    • 52. Mehndiratta S, Chiang YF, Lai MJ et al. Concise syntheses of 7-anilino-indoline-N-benzenesulfonamides as antimitotic and vascular disrupting agents. Bioorg. Med. Chem. 22(17), 4917–4923 (2014).
    • 53. Basha Shaik A. Design and synthesis of pyrazoleeoxindole conjugates targeting tubulin polymerization as new anticancer agents. Eur. J. Med. Chem. 92, 501–513 (2015).
    • 54. Yan J, Hu J, An B, Huang L, Li X. Design, synthesis, and biological evaluation of cyclic-indole derivatives as anti-tumor agents via the inhibition of tubulin polymerization. Eur. J. Med. Chem. 125, 663–675 (2017).
    • 55. Devambatla RKV, Li W, Zaware N et al. Design, synthesis, and structure–activity relationships of pyrimido[4,5-b]indole-4-amines as microtubule depolymerizing agents that are effective against multidrug resistant cells. Bioorg. Med. Chem. Lett. 27(15), 3423–3430 (2017).
    • 56. Ty N, Dupeyre G, Chabot GG et al. Structure-activity relationships of indole compounds derived from combretastatin A4: synthesis and biological screening of 5-phenylpyrrolo[3,4-a]carbazole-1,3-diones as potential antivascular agents. Eur. J. Med. Chem. 45(9), 3726–3739 (2010).
    • 57. Gangjee A, Zaware N, Devambatla RK et al. Synthesis of N(4)-(substituted phenyl)-N(4)-alkyl/desalkyl-9H-pyrimido[4,5-b]indole-2,4-diamines and identification of new microtubule disrupting compounds that are effective against multidrug resistant cells. Bioorg. Med. Chem. 21(4), 891–902 (2013).
    • 58. Parrino B, Carbone A, Ciancimino C et al. Water-soluble isoindolo[2,1-a]quinoxalin-6-imines: in vitro antiproliferative activity and molecular mechanism(s) of action. Eur. J. Med. Chem. 94, 149–162 (2015).
    • 59. Álvarez R, Gajate C, Puebla P, Mollinedo F, Medarde M, Peláez R. Substitution at the indole 3 position yields highly potent indolecombretastatins against human tumor cells. Eur. J. Med. Chem. 158, 167–183 (2018).
    • 60. Spallarossa A, Caneva C, Caviglia M et al. Unconventional Knoevenagel-type indoles: synthesis and cell-based studies for the identification of pro-apoptotic agents. Eur. J. Med. Chem. 102, 648–660 (2015).
    • 61. Wen Z, Xu J, Wang Z et al. 3-(3,4,5-Trimethoxyphenylselenyl)-1H-indoles and their selenoxides as combretastatin A-4 analogs: microwave-assisted synthesis and biological evaluation. Eur. J. Med. Chem. 90, 184–194 (2015).
    • 62. La Regina G, Bai R, Coluccia A et al. New indole tubulin assembly inhibitors cause stable arrest of mitotic progression, enhanced stimulation of natural killer cell cytotoxic activity, and repression of hedgehog-dependent cancer. J. Med. Chem. 58(15), 5789–5807 (2015).
    • 63. Trabbic CJ, George SM, Alexander EM et al. Synthesis and biological evaluation of isomeric methoxy substitutions on anti-cancer indolyl-pyridinyl-propenones: effects on potency and mode of activity. Eur. J. Med. Chem. 122, 79–91 (2016).
    • 64. Romagnoli R, Prencipe F, Oliva P et al. Design, synthesis and biological evaluation of 2-alkoxycarbonyl-3-anilinoindoles as a new class of potent inhibitors of tubulin polymerization. Bioorg. Chem. 97, 103665 (2020).
    • 65. Baytas SN, Inceler N, Yilmaz A et al. Synthesis, biological evaluation and molecular docking studies of trans-indole-3-acrylamide derivatives, a new class of tubulin polymerization inhibitors. Bioorg. Med. Chem. 22(12), 3096–3104 (2014).
    • 66. Naaz F, Ahmad F, Lone BA et al. Design and synthesis of newer 1,3,4-oxadiazole and 1,2,4-triazole based Topsentin analogues as anti-proliferative agent targeting tubulin. Bioorg. Chem. 95, 103519 (2020).
    • 67. Ramya PV Sri, Angapelly S, Guntuku L et al. Synthesis and biological evaluation of curcumin inspired indole analogues as tubulin polymerization inhibitors. Eur. J. Med. Chem. 127, 100–114 (2017).
    • 68. Ghinet A, Abuhaie CM, Gautret P et al. Studies on indolizines. Evaluation of their biological properties as microtubule-interacting agents and as melanoma targeting compounds. Eur. J. Med. Chem. 89, 115–127 (2015).
    • 69. Li W, Shuai W, Sun H et al. Design, synthesis and biological evaluation of quinoline-indole derivatives as anti-tubulin agents targeting the colchicine binding site. Eur. J. Med. Chem. 163, 428–442 (2019).
    • 70. Li W, Sun H, Xu F et al. Synthesis, molecular properties prediction and biological evaluation of indole-vinyl sulfone derivatives as novel tubulin polymerization inhibitors targeting the colchicine binding site. Bioorg. Chem. 85, 49–59 (2019).
    • 71. Lee HY, Pan SL, Su MC et al. Furanylazaindoles: potent anticancer agents in vitro and in vivo. J. Med. Chem. 56(20), 8008–8018 (2013).