364
Views
1
CrossRef citations to date
0
Altmetric
Review

An Update on the Recent Advances and Discovery of Novel Tubulin Colchicine Binding Inhibitors

, , , , & ORCID Icon
Pages 73-95 | Received 31 Aug 2022, Accepted 18 Jan 2023, Published online: 09 Feb 2023

References

  • Siegel RL , MillerKD , FuchsHE , JemalA. Cancer statistics. CA Cancer J. Clin.72(1), 7–33 (2022).
  • Sung H , FerlayJ , SiegelRLet al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer. J. Clin.71(3), 209–249 (2021).
  • Howard J , HymanAA. Dynamics and mechanics of the microtubule plus end. Nature.17,422(6933), 753–8 (2003).
  • Jordan MA , WilsonL. Microtubules as a target for anticancer drugs. Nat. Rev. Cancer.4(4), 253–65 (2004).
  • Downing KH , NogalesE. Tubulin structure: insights into microtubule properties and functions. Curr. Opin. Struct. Biol.8(6), 785–91 (1999).
  • Ferrara R , PilottoS , PerettiUet al. Tubulin inhibitors in non-small cell lung cancer: looking back and forward. Expert. Opin. Pharmacother.17(8), 1113–29 (2016).
  • Chaudhary V , VenghateriJB , DhakedHPSet al. Novel combretastatin-2-aminoimidazole analogues as potent tubulin assembly inhibitors: exploration of unique pharmacophoric impact of bridging skeleton and aryl moiety. J. Med. Chem.59(7), 3439–51 (2016).
  • Steinmetz MO , ProtaAE. Microtubule-targeting agents: strategies to hijack the cytoskeleton. Trends. Cell. Biol.28(10), 776–792 (2018).
  • Dumontet C , JordanMA. Microtubule-binding agents: a dynamic field of cancer therapeutics. Nat. Rev. Drug Discov.9(10), 790–803 (2010).
  • Baytas SN . Recent advances in combretastatin A-4 inspired inhibitors of tubulin polymerization: an update. Curr. Med. Chem.29(20), 3557–3585 (2022).
  • Borisy G , HealdR , HowardJet al. Microtubules: 50 years on from the discovery of tubulin. Nat. Rev. Mol. Cell. Biol.17(5), 322–8 (2016).
  • Ravelli RBG , GigantB , CurmiPAet al. Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature.11, 428(6979), 198–202 (2004).
  • Chinen T , LiuP , ShiodaetSet al. The γ-tubulin-specific inhibitor gatastatin reveals temporal requirements of microtubule nucleation during the cell cycle. Nat. Commun.6, 8722 (2015).
  • Bai R , CovellDG , PeiXFet al. Mapping the binding site of colchicinoids on beta-tubulin. 2-Chloroacetyl-2-demethylthiocolchicine covalently reacts predominantly with cysteine 239 and secondarily with cysteine 354. J. Biol. Chem.275(51), 40443–52 (2000).
  • Kuppens IELM , WitteveenPO , SchotMet al. Phase I dose-finding and pharmacokinetic trial of orally administered indibulin (D-24851) to patients with solid tumors. Invest. New Drugs.25(3), 227–35 (2007).
  • Patterson DM , ZweifelM , MiddletonMRet al. Phase I clinical and pharmacokinetic evaluation of the vascular-disrupting agent OXi4503 in patients with advanced solid tumors. Clin. Cancer Res.18(5), 1415–25 (2012).
  • Liu P , QinY , WuLet al. A phase I clinical trial assessing the safety and tolerability of combretastatin A4 phosphate injections. Anticancer Drugs.25(4), 462–71 (2014).
  • Fox E , MarisJM , WidemannBCet al. A phase 1 study of ABT-751, an orally bioavailable tubulin inhibitor, administered daily for 7 days every 21 days in pediatric patients with solid tumors. Clin. Cancer Res.14(4), 1111–5 (2008).
  • Hawash M . Recent advances of tubulin inhibitors targeting the colchicine binding site for cancer therapy. Biomolecules.12(12), 1843 (2022).
  • Guggilapu SD , LalitaG , ReddyTSet al. Synthesis of C5-tethered indolyl-3-glyoxylamide derivatives as tubulin polymerization inhibitors. Eur. J. Med. Chem.128, 1–12 (2017).
  • Mirzaei H , ShokrzadehM , ModanlooMet al. New indole-based chalconoids as tubulin-targeting anti-proliferative agents. Bioorg. Chem.75, 86–98 (2017).
  • Wang G , PengZ , LiY. Synthesis, anticancer activity and molecular modeling studies of novel chalcone derivatives containing indole and naphthalene moieties as tubulin polymerization inhibitors. Chem. Pharm. Bull.67(7), 725–728 (2019).
  • Kode J , KovvuriJ , NagarajuBet al. Synthesis, biological evaluation, and molecular docking analysis of phenstatin based indole linked chalcones as anticancer agents and tubulin polymerization inhibitors. Bioorg. Chem.105, 104447 (2020).
  • Zhu H , SunH , LiuYet al. Design, synthesis and biological evaluation of vinyl selenone derivatives as novel microtubule polymerization inhibitors. Eur. J. Med. Chem.207, 112716 (2020).
  • Ramya PVS , AngapellyS , GuntukuLet al. Synthesis and biological evaluation of curcumin inspired indole analogues as tubulin polymerization inhibitors. Eur. J. Med. Chem.127, 100–114 (2017).
  • Arnst KE , WangY , HwangDJet al. A potent, metabolically stable tubulin inhibitor targets the colchicine binding site and overcomes taxane resistance. Cancer Res.78(1), 265–277 (2018).
  • Li W , SunH , XuFet 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).
  • Regina GL , BaiR , ColucciaAet 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).
  • Sigalapalli DK , PooladandaV , SinghPet al. Discovery of certain benzyl/phenethyl thiazolidinone-indole hybrids as potential anti-proliferative agents: synthesis, molecular modeling and tubulin polymerization inhibition study. Bioorg. Chem.92, 103188 (2019).
  • Wang Q , ArnstKE , WangYet al. Structure-guided design, synthesis, and biological evaluation of (2-(1h-indol-3-yl)-1h-imidazol-4-yl)(3,4,5-trimethoxyphenyl) methanone (abi-231) analogues targeting the colchicine binding site in tubulin. J. Med. Chem.62(14), 6734–6750 (2019).
  • Cui Y , MaC , ZhangCet al. The discovery of novel indazole derivatives as tubulin colchicine site binding agents that displayed potent antitumor activity both in vitro and in vivo. Eur. J. Med. Chem.187, 111968 (2020).
  • Romagnoli R , PrencipeF , OlivaPet 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).
  • Naaz F , AhmadF , LoneBAet 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).
  • Li G , WangY , LiLet al. Design, synthesis, and bioevaluation of pyrazolo[1,5-a]pyrimidine derivatives as tubulin polymerization inhibitors targeting the colchicine binding site with potent anticancer activities. Eur. J. Med. Chem.202, 112519 (2020).
  • Wu C , WuJ , HuYet al. Design, synthesis and biological evaluation of indole-based[1,2,4]triazolo[4,3-a] pyridine derivatives as novel microtubule polymerization inhibitors. Eur. J. Med. Chem.223, 113629 (2021).
  • Pecnard S , ProvotO , LevaiqueHet al. Cyclic bridged analogs of isoCA-4: Design, synthesis and biological evaluation. Eur. J. Med. Chem.209, 112873 (2021).
  • Wang G , HeM , LiuWet al. Design, synthesis and biological evaluation of novel 2-phenyl-4,5,6,7-tetrahydro-1H-indole derivatives as potential anticancer agents and tubulin polymerization inhibitors. Arabian J. Chem.15, 103504 (2022).
  • Saruengkhanphasit R , ButkinareeC , OrnnorkNet al. Identification of new 3-phenyl-1H-indole-2-carbohydrazide derivatives and their structure–activity relationships as potent tubulin inhibitors and anticancer agents: a combined in silico, in vitro and synthetic study. Bioorg. Chem.110, 104795 (2021).
  • Hawash M , KahramanDC , OlgacAet al. Design and synthesis of novel substituted indole-acrylamide derivatives and evaluation of their anti-cancer activity as potential tubulin-targeting agents. J. Mol. Struct.1254, 132345 (2022).
  • Thanaussavadate B , NgiwsaraL , LirdprapamongkolKet al. A synthetic 2,3-diarylindole induces microtubule destabilization and G2/M cell cycle arrest in lung cancer cells. Bioorg. Med. Chem. Lett.30(1), 126777 (2020).
  • Wang SY , LiuX , MengLWet al. Discovery of indoline derivatives as anticancer agents via inhibition of tubulin polymerization. Bioorg. Med. Chem. Lett.43, 128095 (2021).
  • Diao PC , JianXE , ChenPet al. Design, synthesis and biological evaluation of novel indole-based oxalamide and aminoacetamide derivatives as tubulin polymerization inhibitors. Bioorg. Med. Chem. Lett.30(2), 126816 (2020).
  • Brel VK , ArtyushinOI , Chuprov-NetochinRNet al. Synthesis and biological evaluation of indolylglyoxylamide bisphosphonates, antimitotic microtubule-targeting derivatives of indibulin with improved aqueous solubility. Bioorg. Med. Chem. Lett.30(23), 127635 (2020).
  • Chen P , ZhuangY , DiaoPet al. Synthesis, biological evaluation, and molecular docking investigation of 3-amidoindoles as potent tubulin polymerization inhibitors. Eur. J. Med. Chem.162, 525–533 (2019).
  • Zhai M , LiuS , GaoMet al. 3,5-Diaryl-1H-pyrazolo[3,4-b]pyridines as potent tubulin polymerization inhibitors: rational design, synthesis and biological evaluation. Eur. J. Med. Chem.168, 426–435 (2019).
  • Yin Y , LianBao , XiaYet al. Design, synthesis and biological evaluation of resveratrol-cinnamoyl derivates as tubulin polymerization inhibitors targeting the colchicine binding site. Bioorg. Chem.93, 103319 (2019).
  • Jiang J , ZhangH , WangCet al. 1-Phenyl-dihydrobenzoindazoles as novel colchicine site inhibitors: structural basis and antitumor efficacy. Eur. J. Med. Chem.177, 448–456 (2019).
  • Chen H , DengS , WangYet al. Structure-activity relationship study of novel 6-aryl-2-benzoylpyridines as tubulin polymerization inhibitors with potent antiproliferative properties. J. Med. Chem.63(2), 827–846 (2020).
  • Li L , QuanD , ChenJet al. Design, synthesis, and biological evaluation of 1-substituted -2-aryl imidazoles targeting tubulin polymerization as potential anticancer agents. Eur. J. Med. Chem.184, 111732 (2019).
  • Zhao W , HeL , XiangT , TangY. Discover 4b-NH-(6-aminoindole)-4-desoxy-podophyllotoxin with nanomolar-potency antitumor activity by improving the tubulin binding affinity on the basis of a potential binding site nearby colchicine domain. Eur. J. Med. Chem.170, 73–86 (2019).
  • Song J , GaoQ , WuBet al. Discovery of tertiary amide derivatives incorporating benzothiazole moiety as anti-gastric cancer agents in vitro via inhibiting tubulin polymerization and activating the Hippo signaling pathway. Eur. J. Med. Chem.203, 112618 (2020).
  • Sun Y , SongJ , KongLet al. Design, synthesis and evaluation of novel bis-substituted aromatic amide dithiocarbamate derivatives as colchicine site tubulin polymerization inhibitors with potent anticancer activities. Eur. J. Med. Chem.229, 114069 (2022).
  • Liu X , PangX , LiuYet al. Discovery of novel diarylamide n-containing heterocyclic derivatives as new tubulin polymerization inhibitors with anti-cancer activity. Molecules.26(13), 4047 (2021).
  • Yang B , ZhouJ , WangFet al. Pyrazoline derivatives as tubulin polymerization inhibitors with one hit for vascular endothelial growth factor receptor 2 inhibition. Bioorg. Chem.114, 105134 (2021).
  • Ren Y , WangY , LiGet al. Discovery of novel benzimidazole and indazole analogues as tubulin polymerization inhibitors with potent anticancer activities. J. Med. Chem.64, 4498–4515 (2021).
  • Chen H , DengS , AlbadariNet al. Design, synthesis, and biological evaluation of stable colchicine binding site tubulin inhibitors 6-aryl-2-benzoyl-pyridines as potential anticancer agents. J. Med. Chem.64(16), 12049–12074 (2021).
  • Yan X , LengJ , ChenTet al. Design, synthesis, and biological evaluation of novel diphenylamine derivatives as tubulin polymerization inhibitors targeting the colchicine binding site. Eur. J. Med. Chem.237, 114372 (2022).
  • Li W , ShuaiW , XuFet al. Discovery of novel 4-arylisochromenes as anticancer agents inhibiting tubulin polymerization. ACS Med. Chem. Lett.9(10), 974–979 (2018).
  • Ansari M , ShokrzadehM , KarimaSet al. New thiazole-2(3H)-thiones containing 4-(3,4,5-trimethoxyphenyl) moiety as anticancer agents. Eur. J. Med. Chem.185, 111784 (2020).
  • He JP , ZhangM , TangLet al. Synthesis, biological evaluation, and molecular docking of arylpyridines as antiproliferative agent targeting tubulin. ACS Med. Chem. Lett.11(8), 1611–1619 (2020).
  • Lai Q , WangY , WangRet al. Design, synthesis and biological evaluation of a novel tubulin inhibitor 7a3 targeting the colchicine binding site. Eur. J. Med. Chem.156, 162–179 (2020).
  • Hawash M , QaoudMT , JaradatNet al. Anticancer activity of thiophene carboxamide derivatives as CA-4 biomimetics: synthesis, biological potency, 3D spheroid model, and molecular dynamics simulation. Biomimetics.7(4), 247 (2022).
  • Xu Q , BaoK , SunMet al. Design, synthesis and structureactivity relationship of 3,6-diaryl-7H-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazines as novel tubulin inhibitors. Sci Rep.7(1), 11997 (2017).
  • Fu D , LiP , WuBet al. Molecular diversity of trimethoxyphenyl-1,2,3-triazole hybrids as novel colchicine site tubulin polymerization inhibitors. Eur. J. Med. Chem.165, 309–322 (2019).
  • Qi Z , HaoS , TianHet al. Synthesis and biological evaluation of 1-(benzofuran-3-yl)-4-(3,4,5-tr-imethoxyphenyl)-1H-1,2,3-triazole derivatives as tubulin polymerization inhibitors. Bioorg. Chem.94, 103392 (2020).
  • Mustafa M , AnwarS , ElgamalFet al. Potent combretastatin A-4 analogs containing 1,2,4-triazole: synthesis, antiproliferative, anti-tubulin activity, and docking study. Eur. J. Med. Chem.183, 111697 (2019).
  • Ansaria M , ShokrzadehM , KarimaSet al. Design, synthesis and biological evaluation of flexible and rigid analogs of 4H-1,2,4-triazoles bearing 3,4,5-trimethoxyphenyl moiety as new antiproliferative agents. Bioorg. Chem.93, 103300 (2019).
  • Wang C , LiY , LiuTet al. Design, synthesis and evaluation of antiproliferative and antitubulin activities of 5-methyl-4-aryl-3-(4-arylpiperazine-1-carbonyl)-4H-1,2,4-triazoles. Bioorg. Chem.104, 103909 (2020).
  • Hua S , ChenF , WangX , GouS. Dual-functional conjugates improving cancer immunochemotherapy by inhibiting tubulin polymerization and indoleamine-2,3-dioxygenase. Eur. J. Med. Chem.189, 112041 (2020).
  • Yang F , JianX , DiaoPet al. Synthesis, and biological evaluation of 3,6-diaryl-[1,2,4]triazolo[4,3-a]pyridine analogues as new potent tubulin polymerization inhibitors. Eur. J. Med. Chem.204, 112625 (2020).
  • Huo X , JianX , YangJet al. Discovery of highly potent tubulin polymerization inhibitors: design, synthesis, and structure-activity relationships of novel 2,7-diaryl-[1,2,4]triazolo[1,5-a]pyrimidines. Eur. J. Med. Chem.220, 113449 (2021).
  • Eissa IH , DahabMA , IbrahimetMKet al. Design and discovery of new antiproliferative 1,2,4-triazin-3(2H)-ones as tubulin polymerization inhibitors targeting colchicine binding site. Bioorg. Chem.112, 104965 (2021).
  • Wu M , ManR , LiaoYet al. Discovery of novel indole-1,2,4-triazole derivatives as tubulin polymerization inhibitors. Drug. Dev Res.82(7), 1008–1020 (2021).
  • Mohamed HS , AminNH , El-SaadiMT , Abdel-RahmanHM. Design, synthesis, biological assessment, and in silico studies of 1,2,4-triazolo[1,5-a]pyrimidine derivatives as tubulin polymerization inhibitors. Bioorg. Chem.121, 105687 (2022).
  • Ma W , ChenP , HuoXet al. Development of triazolothiadiazine derivatives as highly potent tubulin polymerization inhibitors: structure-activity relationship, in vitro and in vivo study. Eur. J. Med. Chem.208, 112847 (2020).
  • Yang F , YuLZ , DiaoPCet al. Novel [1,2,4]triazolo[1,5-a]pyrimidine derivatives as potent antitubulin agents: design, multicomponent synthesis and antiproliferative activities. Bioorg. Chem.92, 103260 (2019).
  • Yang F , HeCP , DiaoPCet al. Discovery and optimization of 3,4,5-trimethoxyphenyl substituted triazolylthioacetamides as potent tubulin polymerization inhibitors. Bioorg. Med. Chem. Lett.29(1), 22–27 (2019).
  • Mustafa M , AbdelhamidD , AbdelhafezEMNet al. Synthesis, antiproliferative, anti-tubulin activity, and docking study of new 1,2,4-triazoles as potential combretastatin analogues. Eur. J. Med. Chem.141, 293–305 (2017).
  • Li W , YinY , ShuaiWet al. Discovery of novel quinazolines as potential anti-tubulin agents occupying three zones of colchicine domain. Bioorg. Chem.83, 380–390 (2019).
  • Liang T , ZhouX , LuLet al. Structure-activity relationships and antiproliferative effects of 1,2,3,4-4H-quinoxaline derivatives as tubulin polymerization inhibitors. Bioorg. Chem.110, 104793 (2021).
  • Ren Y , RuanY , ChengBet al. Design, synthesis and biological evaluation of novel acridine and quinoline derivatives as tubulin polymerization inhibitors with anticancer activities. Bioorg. Med. Chem.46, 116376 (2021).
  • Wang K , ZhongH , LiNet al. Discovery of novel anti-breast-cancer inhibitors by synergistically antagonizing microtubule polymerization and aryl hydrocarbon receptor expression. J. Med. Chem.64(17), 12964–12977 (2021).
  • Li W , ShuaiW , SunHet 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).
  • Elhemely MA , BelgathAA , El-SayedSet al. SAR of novel 3-arylisoquinolinones: meta-substitution on the aryl ring dramatically enhances antiproliferative activity through binding to microtubules. J. Med. Chem.65(6), 4783–4797 (2022).
  • Li W , XuF , ShuaiWet al. Discovery of novel quinoline-chalcone derivatives as potent antitumor agents with microtubule polymerization inhibitory activity. J. Med. Chem.62(2), 993–1013 (2019).
  • Mirzaei S , EisvandF , HadizadehFet al. Design, synthesis and biological evaluation of novel 5,6,7-trimethoxy-N-aryl-2-styrylquinolin-4-amines as potential anticancer agents and tubulin polymerization inhibitors. Bioorg. Chem.98, 103711 (2020).
  • Rehulka J , VychodilovaK , KrejcíPet al. Fluorinated derivatives of 2-phenyl-3-hydroxy-4(1H)-quinolinone as tubulin polymerization inhibitors. Eur. J. Med. Chem.192, 112176 (2020).
  • Khelifi I , NaretT , HamzeAet al. N,N-bis-heteroaryl methylamines: potent anti-mitotic and highly cytotoxic agents. Eur. J. Med. Chem.168, 176–188 (2019).
  • Bueno O , GargantillaM , Estévez-GallegoetJet al. Diphenyl ether derivatives occupy the expanded binding site of cyclohexanedione compounds at the colchicine site in tubulin by movement of the αT5 loop. Eur. J. Med. Chem.171, 195–208 (2019).
  • Hu X , LiL , ZhangQet al. Design, synthesis and biological evaluation of a novel tubulin inhibitor SKLB0565 targeting the colchicine binding site. Bioorg. Chem.97, 103695 (2020).
  • Shao Y , YinY , LianBet al. Synthesis and biological evaluation of novel shikonin-benzo[b]furanderivatives as tubulin polymerization inhibitors targeting the colchicine binding site. Eur. J. Med. Chem.190, 112105 (2020).
  • Wang G , LiuW , TangJet al. Design, synthesis, and anticancer evaluation of benzophenone derivatives bearing naphthalene moiety as novel tubulin polymerization inhibitors. Bioorg. Chem.104, 104265 (2020).
  • Liu R , ZhangS , HuangMet al. Design, synthesis and bioevaluation of 2,7-diaryl-pyrazolo[1,5-a] pyrimidines as tubulin polymerization inhibitors. Bioorg. Chem.115, 105220 (2021).
  • Zhu H , LiW , ShuaiWet al. Discovery of novel N-benzylbenzamide derivatives as tubulin polymerization inhibitors with potent antitumor activities. Eur. J. Med. Chem.216, 113316 (2021).
  • Cheng B , ZhuG , MengLet al. Identification and optimization of biphenyl derivatives as novel tubulin inhibitors targeting colchicine-binding site overcoming multidrug resistance. Eur. J. Med. Chem.228, 113930 (2022).
  • Su C , MingQ , RahmanKet al. Salvia miltiorrhiza: traditional medicinal uses, chemistry, and pharmacology. Chin. J. Nat. Med.13(3), 163–82 (2015).
  • Zhang Y , JiangP , YeMet al. Tanshinones: sources, pharmacokinetics and anti-cancer activities. Int. J. Mol. Sci.13(10), 13621–66 (2012).
  • Huang H , YaoY , HouGet al. Design, synthesis and biological evaluation of tanshinone IIA-based analogues: potent inhibitors of microtubule formation and angiogenesis. Eur. J. Med. Chem.224, 113708 (2021).
  • Xu F , LiW , ShuaiWet al. Design, synthesis and biological evaluation of pyridine-chalcone derivatives as novel microtubule-destabilizing agents. Eur. J. Med. Chem.173, 1–14 (2019).
  • Lin Y , TungpraditR , SinchaikulSet al. Targeting the delivery of glycan-based paclitaxel prodrugs to cancer cells via glucose transporters. J. Med. Chem.51(23), 7428–41 (2008).
  • Calvaresi EC , HergenrotherPJ. Glucose conjugation for the specific targeting and treatment of cancer. Chem. Sci.4(6), 2319–2333 (2013).
  • Liu P , LuY , GaoXet al. Highly water-soluble platinum (II) complexes as GLUT substrates for targeted therapy: improved anticancer efficacy and transportermediated cytotoxic properties. Chem. Commun.49(24), 2421–3 (2013).
  • Han J , GaoX , LiuRet al. Design, synthesis of novel platinum (II) glycoconjugates, and evalution of their antitumor effects. Chem. Biol. Drug Des.87(6), 867–77 (2016).
  • Wang Z , LiuR , ZhangXet al. Design, synthesis and biological evaluation of colchicine glycoconjugates as tubulin polymerization inhibitors. Bioorg. Med. Chem.58, 116671 (2022).
  • Zheng Y , DongY , SiS , ZhenY , GongJ. IMB5476, a novel microtubule inhibitor, induces mitotic catastrophe and overcomes multidrug resistance in tumors. Eur. J. Pharmacol.919, 174802 (2022).
  • Shuai W , LiX , LiWet al. Design, synthesis and anticancer properties of isocombretapyridines as potent colchicine binding site inhibitors. Eur. J. Med. Chem.197, 112308 (2020).
  • Kumar B , SharmaP , GuptaVPet al. Synthesis and biological evaluation of pyrimidine bridged combretastatin derivatives as potential anticancer agents and mechanistic studies. Bioorg. Chem.78, 130–140 (2018).
  • Liu Y , WuY , SunLet al. Synthesis and structure-activity relationship study of water-soluble carbazole sulfonamide derivatives as new anticancer agents. Eur. J. Med. Chem.191, 112181 (2020).
  • Shaheen MA , El-EmamAA , El-GoharyNS. 1,4,5,6,7,8-Hexahydroquinolines and 5,6,7,8-tetrahydronaphthalenes: a new class of antitumor agents targeting the colchicine binding site of tubulin. Bioorg. Chem.99, 103831 (2020).
  • Shuai W , WangG , ZhangYet al. Recent progress on tubulin inhibitors with dual targeting capabilities for cancer therapy. J. Med. Chem.64(12), 7963–7990 (2021).
  • Zhu H , TanY , HeCet al. Discovery of a novel vascular disrupting agent inhibiting tubulin polymerization and HDACs with potent antitumor effects. J. Med. Chem.65(16), 11187–11213 (2022).
  • Fu D , WangT. Discovery of dual tubulin-NEDDylation inhibitors with antiproliferative activity. J. Enzyme Inhib. Med. Chem.38(1), 166–175 (2022).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.