Acessibilidade / Reportar erro

Natural products as new antimitotic compounds for anticancer drug development

Abstract

Cell cycle control genes are frequently mutated in cancer cells, which usually display higher rates of proliferation than normal cells. Dysregulated mitosis leads to genomic instability, which contributes to tumor progression and aggressiveness. Many drugs that disrupt mitosis have been studied because they induce cell cycle arrest and tumor cell death. These antitumor compounds are referred to as antimitotics. Vinca alkaloids and taxanes are natural products that target microtubules and inhibit mitosis, and their derivatives are among the most commonly used drugs in cancer therapy worldwide. However, severe adverse effects such as neuropathies are frequently observed during treatment with microtubule-targeting agents. Many efforts have been directed at developing improved antimitotics with increased specificity and decreased likelihood of inducing side effects. These new drugs generally target specific components of mitotic regulation that are mainly or exclusively expressed during cell division, such as kinases, motor proteins and multiprotein complexes. Such small molecules are now in preclinical studies and clinical trials, and many are products or derivatives from natural sources. In this review, we focused on the most promising targets for the development of antimitotics and discussed the advantages and disadvantages of these targets. We also highlighted the novel natural antimitotic agents under investigation by our research group, including combretastatins, withanolides and pterocarpans, which show the potential to circumvent the main issues in antimitotic therapy.

Antimitotic Agents; Microtubules; Spindle Apparatus; Mitosis; Cancer


INTRODUCTION

Cancer pathogenesis is a complex process in which normal cycling cells acquire basic neoplastic characteristics typically common to all tumor types. Cells from normal tissues exhibit growth entirely controlled by the cell division machinery, which is orchestrated by the production, release, recognition and inhibition of growth-promoting signals to foster tissue homeostasis. Tumor cells escape from this growth orchestration through various pathways, usually those linked to self-sufficient growth. Therefore, an essential feature of cancer cells is the ability to maintain chronically disrupted homeostatic proliferation (11. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74, http://dx.doi.org/10.1016/j.cell.2011.02.013.
http://dx.doi.org/10.1016/j.cell.2011.02...
). Mitosis is the fundamental proliferation process of division in human somatic cells; dysregulated mitosis leads to genomic instability characterized by DNA mutations and chromosomal aberrations. These genetic changes result in proliferative advantages for neoplastic cells and increase the susceptibility of these cells to the accumulation of additional genetic mutations, which in turn contribute to tumor progression and an exacerbated aggressive phenotype (22. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9(3):153-66, http://dx.doi.org/10.1038/nrc2602.
http://dx.doi.org/10.1038/nrc2602...
).

Some compounds used in cancer chemotherapy target protein components of the mitotic regulatory machinery, acting as cell division blockers and, consequently, cell death inducers (33. Marzo I, Naval J. Antimitotic drugs in cancer chemotherapy: promises and pitfalls. Biochem Pharmacol. 2013;86(6):703-10, http://dx.doi.org/10.1016/j.bcp.2013.07.010.
http://dx.doi.org/10.1016/j.bcp.2013.07....
). Many proteins involved in this process play an essential role in mitosis, making them interesting targets for the development of antimitotic anticancer drugs. The clinical success of compounds that disrupt microtubule dynamics, such as vinca alkaloids and taxanes, stimulated the development of new drugs with specific mitotic targets (44. Sudakin V, Yen TJ. Targeting mitosis for anti-cancer therapy. BioDrugs. 2007;21(4):225-33, http://dx.doi.org/10.2165/00063030-200721040-00003.
http://dx.doi.org/10.2165/00063030-20072...
). Substantial research aimed at the development of new antimitotic compounds with different targets and increasingly selective antitumor activity has ensued. Herein, we discuss traditional and novel antimitotics with antitumor potential and the respective cellular targets of these agents, as summarized in Figure 1.

Figure 1
Antimitotic targets and their respective inhibitors as cited in the text. The targets are placed in the illustration according to the cell cycle phase in which they perform their main functions. The corresponding inhibitors are listed in the legend. The phases of mitosis are depicted in blue, the phases of interphase are in green, and cell cycle checkpoints are in red. The compounds (+)2,3,9-trimethoxypterocarpan and withaphysalin F are not included, because their targets are unknown.

Antimitotic agents and their main targets

Mitosis is a crucial phase of the cell cycle when the chromosomes must be aligned on the metaphase plate and segregated correctly to generate two identical daughter cells. The search for new clinically effective antitumor compounds has yielded many drugs directed at targets expressed during mitosis. Such compounds, called antimitotics, target proteins involved in this complex process and are mitotic arrest agents (55. Penna LS, Henriques JAP, Bonatto D. Anti-mitotic agents: are they emerging molecules for cancer treatment? Pharmacol Ther. 2017;173:67-82, http://dx.doi.org/10.1016/j.pharmthera.2017.02.007.
http://dx.doi.org/10.1016/j.pharmthera.2...
).

Microtubules and microtubule-associated proteins

Microtubules are composed of α- and β-tubulin heterodimers that rapidly polymerize and depolymerize. Many microtubule-associated proteins (MAPs) fine-tune the organization of microtubules and promote or suppress microtubule dynamics (66. Muroyama A, Lechler T. Microtubule organization, dynamics and functions in differentiated cells. Development. 2017;144(17):3012-21, http://dx.doi.org/10.1242/dev.153171.
http://dx.doi.org/10.1242/dev.153171...
). Microtubules play important roles in a wide range of cellular functions, such as the assembly of the mitotic spindle; the movement of organelles, vesicles and proteins; and the associated cell signaling, as well as in the development and maintenance of cellular shape (77. Harrison MR, Holen KD, Liu G. Beyond taxanes: a review of novel agents that target mitotic tubulin and microtubules, kinases, and kinesins. Clin Adv Hematol Oncol. 2009;7(1):54-64.). Chromosome segregation is first enabled by the connection between mitotic spindle microtubules and the kinetochore, a macromolecular complex positioned at the chromosomal centromere, which is assembled at the beginning of mitosis (44. Sudakin V, Yen TJ. Targeting mitosis for anti-cancer therapy. BioDrugs. 2007;21(4):225-33, http://dx.doi.org/10.2165/00063030-200721040-00003.
http://dx.doi.org/10.2165/00063030-20072...
). Coupling between microtubules and the kinetochore is essential for correct spindle formation and subsequent chromosome alignment and segregation (66. Muroyama A, Lechler T. Microtubule organization, dynamics and functions in differentiated cells. Development. 2017;144(17):3012-21, http://dx.doi.org/10.1242/dev.153171.
http://dx.doi.org/10.1242/dev.153171...
). Unsurprisingly, the first antimitotic drugs developed interfere with microtubule dynamics, through either the inhibition or stabilization of microtubule polymerization.

Tubulin-targeting antimitotics bind tubulin at different sites and may exert different effects. Vinca alkaloids (vincristine, vinblastine, vindesine, vinorelbine and vinflunine) are natural products capable of binding and destabilizing the polymeric structure of microtubules, thus causing microtubule disassembly. Colchicine also acts via this mechanism but at another interaction site. Taxanes (paclitaxel and docetaxel) and epothilones are examples of microtubule-stabilizing agents that prevent depolymerization (88. Campos SM, Dizon DS. Antimitotic inhibitors. Hematol Oncol Clin North Am. 2012;26(3):607-28, http://dx.doi.org/10.1016/j.hoc.2012.01.007.
http://dx.doi.org/10.1016/j.hoc.2012.01....
). Tubulin-binding agents traditionally used in the clinic kill dividing tumor and healthy cells and affect microtubule dynamics in nonproliferating cells, resulting in various side effects, such as myelosuppression and neuropathies (99. Gascoigne KE, Taylor SS. How do anti-mitotic drugs kill cancer cells? J Cell Sci. 2009;122(Pt 15):2579-85, http://dx.doi.org/10.1242/jcs.039719.
http://dx.doi.org/10.1242/jcs.039719...
). Nevertheless, tubulin-binding agents are widely employed in the treatment of several solid tumors and oncohematological malignancies (88. Campos SM, Dizon DS. Antimitotic inhibitors. Hematol Oncol Clin North Am. 2012;26(3):607-28, http://dx.doi.org/10.1016/j.hoc.2012.01.007.
http://dx.doi.org/10.1016/j.hoc.2012.01....
).

After the first tubulin-binding agents showed antitumor effects, many other tubulin-targeting antimitotic drugs were developed, along with related inhibitors directed at other targets such as kinases and mitotic protein complexes, aimed at inhibiting cell proliferation (55. Penna LS, Henriques JAP, Bonatto D. Anti-mitotic agents: are they emerging molecules for cancer treatment? Pharmacol Ther. 2017;173:67-82, http://dx.doi.org/10.1016/j.pharmthera.2017.02.007.
http://dx.doi.org/10.1016/j.pharmthera.2...
). Among the MAPs, the kinesin spindle protein (KSP) Eg5 is a kinesin responsible for the separation of centrosomes and for the bipolar configuration of the mitotic spindle. Inhibition of Eg5 causes monopolar spindle formation and cell cycle arrest, an effect demonstrated by monastrol, a small-molecule Eg5 inhibitor (1010. Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL, Mitchison TJ. Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science. 1999;286(5441):971-4, http://dx.doi.org/10.1126/science.286.5441.971.
http://dx.doi.org/10.1126/science.286.54...
,1111. Jackson JR, Patrick DR, Dar MM, Huang PS. Targeted anti-mitotic therapies: can we improve on tubulin agents? Nat Rev Cancer. 2007;7(2):107-17, http://dx.doi.org/10.1038/nrc2049.
http://dx.doi.org/10.1038/nrc2049...
). Other Eg5 inhibitors, such as ispinesib (Cytokinetics), SB743921 (Cytokinetics), AZD4877 (AstraZeneca), Arq621 (ArQule), EMD-534085 (Merck-K GaA), MK-0731 (Merck & Co.), filanesib (Array Biopharma) and litronesib (Kyowa Hakko Kirin/Eli Lilly), have reached clinical trials (1212. Myers SM, Collins I. Recent findings and future directions for interpolar mitotic kinesin inhibitors in cancer therapy. Future Med Chem. 2016;8(4):463-89, http://dx.doi.org/10.4155/fmc.16.5.
http://dx.doi.org/10.4155/fmc.16.5...
). Centromeric protein E (CENP-E) is another MAP inhibited by antitumor agents. CENP-E is essential for the alignment of chromosomes during metaphase and passage to anaphase (33. Marzo I, Naval J. Antimitotic drugs in cancer chemotherapy: promises and pitfalls. Biochem Pharmacol. 2013;86(6):703-10, http://dx.doi.org/10.1016/j.bcp.2013.07.010.
http://dx.doi.org/10.1016/j.bcp.2013.07....
). Examples of CENP-E inhibitors are PF-2771 (1313. Kung PP, Martinez R, Zhu Z, Zager M, Blasina A, Rymer I, et al. Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer. Mol Cancer Ther. 2014;13(8):2104-15, http://dx.doi.org/10.1158/1535-7163.MCT-14-0083-T.
http://dx.doi.org/10.1158/1535-7163.MCT-...
), UA62784 (1414. Henderson MC, Shaw YJ, Wang H, Han H, Hurley LH, Flynn G, et al. UA62784, a novel inhibitor of centromere protein E kinesin-like protein. Mol Cancer Ther. 2009;8(1):36-44, http://dx.doi.org/10.1158/1535-7163.MCT-08-0789.
http://dx.doi.org/10.1158/1535-7163.MCT-...
) and Cmpd-A (1515. Ohashi A, Ohori M, Iwai K, Nambu T, Miyamoto M, Kawamoto T, et al. A Novel Time-Dependent CENP-E Inhibitor with Potent Antitumor Activity. PLoS One. 2015;10(12):e0144675, http://dx.doi.org/10.1371/journal.pone.0144675.
http://dx.doi.org/10.1371/journal.pone.0...
), but only GSK923295 has reached clinical trials (1616. Chung V, Heath EI, Schelman WR, Johnson BM, Kirby LC, Lynch KM, et al. First-time-in-human study of GSK923295, a novel antimitotic inhibitor of centromere-associated protein E (CENP-E), in patients with refractory cancer. Cancer Chemother Pharmacol. 2012;69(3):733-41, http://dx.doi.org/10.1007/s00280-011-1756-z.
http://dx.doi.org/10.1007/s00280-011-175...
). Abnormalities in the function and expression of kinesins are important for the development and progression of many human cancers, suggesting that this class of proteins is an interesting target for new anticancer therapy strategies (1717. Hegde PS, Cogswell J, Carrick K, Jackson J, Wood KW, Eng WK, et al. Differential gene expression analysis of kinesin spindle protein in human solid tumors. Proc Am Soc Clin Oncol. 2003;22:535.,1818. Maliga Z, Junqueira M, Toyoda Y, Ettinger A, Mora-Bermúdez F, Klemm RW, et al. A genomic toolkit to investigate kinesin and myosin motor function in cells. Nat Cell Biol. 2013;15(3):325-34, http://dx.doi.org/10.1038/ncb2689.
http://dx.doi.org/10.1038/ncb2689...
). Since some kinesins are expressed only during mitosis, their inhibition may reduce side effects related to antineoplastic agents targeting interphase proteins, such as traditional tubulintargeting antimitotics (99. Gascoigne KE, Taylor SS. How do anti-mitotic drugs kill cancer cells? J Cell Sci. 2009;122(Pt 15):2579-85, http://dx.doi.org/10.1242/jcs.039719.
http://dx.doi.org/10.1242/jcs.039719...
).

Cell cycle regulatory kinases

Mitosis is a strictly controlled set of events driven by many regulatory proteins that ensure cellular modifications suitable for the correct replication and distribution of genomic DNA between daughter cells. Among the proteins targeted by new antitumor agent prototypes are cell cycle regulators such as checkpoint kinases (CHKs) and cyclin-dependent kinases (CDKs), as well as regulators of the mitotic process, including Aurora kinases (AURKs) and polo-like kinases (PLKs) (1919. Pitts TM, Davis SL, Eckhardt SG, Bradshaw-Pierce EL. Targeting nuclear kinases in cancer: development of cell cycle kinase inhibitors. Pharmacol Ther. 2014;142(2):258-69, http://dx.doi.org/10.1016/j.pharmthera.2013.12.010.
http://dx.doi.org/10.1016/j.pharmthera.2...
).

CDKs and CHKs control cell cycle progression. CHKs are responsible for maintaining cell genomic integrity since they are activated in response to DNA damage detected by molecular checking mechanisms in a temporally and spatially controlled manner. Activation of CHK1 and CHK2 results in cell cycle arrest, so that DNA is repaired or, if repair is not possible, apoptosis can be triggered. The defects in checkpoint signaling in many types of cancer are important drivers for the development of new drugs targeting CHKs (2020. Bartek J, Lukas J. Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell. 2003;3(5):421-9, http://dx.doi.org/10.1016/S1535-6108(03)00110-7.
http://dx.doi.org/10.1016/S1535-6108(03)...
). In addition, CDKs are kinases that control crucial steps in the cell cycle and are regulated by binding proteins called cyclins. Gene products that promote cell cycle progression are often mutated and constitutively active in tumor cells, increasing the interest in the development of selective inhibitors of CDKs and mitotic kinases (2121. Ortega S, Malumbres M, Barbacid M. Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta. 2002;1602(1):73-87, http://dx.doi.org/10.1016/S0304-419X(02)00037-9.
http://dx.doi.org/10.1016/S0304-419X(02)...
). CDK inhibitors (CDKIs) include staurosporine, alkaloids and flavonoids, many of which have undergone clinical studies (2222. Cirillo D, Pentimalli F, Giordano A. Peptides or small molecules? Different approaches to develop more effective CDK inhibitors. Curr Med Chem. 2011;18(19):2854-66, http://dx.doi.org/10.2174/092986711796150496.
http://dx.doi.org/10.2174/09298671179615...
,2323. Canavese M, Santo L, Raje N. Cyclin dependent kinases in cancer: potential for therapeutic intervention. Cancer Biol Ther. 2012;13(7):451-7, http://dx.doi.org/10.4161/cbt.19589.
http://dx.doi.org/10.4161/cbt.19589...
24. Roshak AK, Capper EA, Imburgia C, Fornwald J, Scott G, Marshall LA. The human polo-like kinase, PLK, regulates cdc2/cyclin B through phosphorylation and activation of the cdc25C phosphatase. Cell Signal. 2000;12(6):405-11, http://dx.doi.org/10.1016/S0898-6568(00)00080-2.
http://dx.doi.org/10.1016/S0898-6568(00)...
).

PLK1 is the most studied member of the PLK family, with important roles in some cell cycle steps, such as the activation of the CDK1-cyclin B complex, the onset of chromosomal segregation and cytokinesis (2525. Tut TG, Lim SH, Dissanayake IU, Descallar J, Chua W, Ng W, et al. Upregulated Polo-like Kinase 1 expression correlates with inferior survival outcomes in rectal cancer. PLoS One. 2015;10(6):e0129313, http://dx.doi.org/10.1371/journal.pone.0129313.
http://dx.doi.org/10.1371/journal.pone.0...
). The expression of PLK1 is often elevated in tumors, which is correlated with a poor prognosis; thus, PLK1 is a target for anticancer therapy (2626. Kanaji S, Saito H, Tsujitani S, Matsumoto S, Tatebe S, Kondo A, et al. Expression of polo-like kinase 1 (PLK1) protein predicts the survival of patients with gastric carcinoma. Oncology. 2006;70(2):126-33, http://dx.doi.org/10.1159/000093003.
http://dx.doi.org/10.1159/000093003...
). Indeed, the development of PLK inhibitors is currently focused on PLK1 and has resulted in two promising inhibitors in clinical trials, namely, rigosertib and volasertib (2727. Gumireddy K, Reddy MV, Cosenza SC, Boominathan R, Baker SJ, Papathi N, et al. ON01910, a non-ATP-competitive small molecule inhibitor of Plk1, is a potent anticancer agent. Cancer Cell. 2005;7(3):275-86, http://dx.doi.org/10.1016/j.ccr.2005.02.009.
http://dx.doi.org/10.1016/j.ccr.2005.02....
,2828. Rudolph D, Steegmaier M, Hoffmann M, Grauert M, Baum A, Quant J, et al. BI 6727, a Polo-like kinase inhibitor with improved pharmacokinetic profile and broad antitumor activity. Clin Cancer Res. 2009;15(9):3094-102, http://dx.doi.org/10.1158/1078-0432.CCR-08-2445.
http://dx.doi.org/10.1158/1078-0432.CCR-...
).

The Aurora family consists of serine-threonine kinases with a pivotal role in the control of the cell cycle and mitosis. Aurora A is located in the centrosome and acts in the progression from the G1 phase to the S phase of the cell cycle and in the initial stages of mitosis, enabling centrosome separation and mitotic spindle assembly. Aurora B is found in the mitotic spindle and controls chromosome condensation, in addition to acting at the end of mitosis to regulate cytokinesis (2929. Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel therapy targets in cancers. Oncotarget. 2017;8(14):23937-54, http://dx.doi.org/10.18632/oncotarget.14893.
http://dx.doi.org/10.18632/oncotarget.14...
). Both Auroras have an oncogenic role in human tumors (3030. Cirak Y, Furuncuoglu Y, Yapicier O, Aksu A, Cubukcu E. Aurora A overexpression in breast cancer patients induces taxane resistance and results in worse prognosis. J BUON. 2015;20(6):1414-9. 31. Davidson B, Nymoen DA, Elgaaen BV, Staff AC, Tropé CG, Kærn J, et al. BUB1 mRNA is significantly co-expressed with AURKA and AURKB mRNA in advanced-stage ovarian serous carcinoma. Virchows Arch. 2014;464(6):701-7, http://dx.doi.org/10.1007/s00428-014-1577-7.
http://dx.doi.org/10.1007/s00428-014-157...
32. Jeng YM, Peng SY, Lin CY, Hsu HC. Overexpression and amplification of Aurora-A in hepatocellular carcinoma. Clin Cancer Res. 2004;10(6):2065-71, http://dx.doi.org/10.1158/1078-0432.CCR-1057-03.
http://dx.doi.org/10.1158/1078-0432.CCR-...
-3333. Heredia FF, de Sousa JC, Ribeiro Junior HL, Carvalho AF, Magalhães SM, Pinheiro RF. Proteins related to the spindle and checkpoint mitotic emphasize the different pathogenesis of hypoplastic MDS. Leuk Res. 2014;38(2):218-24, http://dx.doi.org/10.1016/j.leukres.2013.11.003.
http://dx.doi.org/10.1016/j.leukres.2013...
). Among the effects associated with tumorigenesis, Aurora A overexpression leads to chromosomal instability characterized by aneuploidy and the premature segregation of sister chromatids (3434. Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov. 2009;8(7):547-66, http://dx.doi.org/10.1038/nrd2907.
http://dx.doi.org/10.1038/nrd2907...
). Inhibition of Aurora kinases is a strategy in cancer treatment, especially in combined therapeutic approaches. Most Aurora inhibitors, such as alisertib, tozasertib and barasertib, compete with ATP for binding to the catalytic site (3535. Matulonis UA, Sharma S, Ghamande S, Gordon MS, Del Prete SA, Ray-Coquard I, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol. 2012;127(1):63-9, http://dx.doi.org/10.1016/j.ygyno.2012.06.040.
http://dx.doi.org/10.1016/j.ygyno.2012.0...
36. Yang J, Ikezoe T, Nishioka C, Tasaka T, Taniguchi A, Kuwayama Y, et al. AZD1152, a novel and selective aurora B kinase inhibitor, induces growth arrest, apoptosis, and sensitization for tubulin depolymerizing agent or topoisomerase II inhibitor in human acute leukemia cells in vitro and in vivo. Blood. 2007;110(6):2034-40, http://dx.doi.org/10.1182/blood-2007-02-073700.
http://dx.doi.org/10.1182/blood-2007-02-...
-3737. Giles FJ, Cortes J, Jones D, Bergstrom D, Kantarjian H, Freedman SJ. MK-0457, a novel kinase inhibitor, is active in patients with chronic myeloid leukemia or acute lymphocytic leukemia with the T315I BCR-ABL mutation. Blood. 2007;109(2):500-2, http://dx.doi.org/10.1182/blood-2006-05-025049.
http://dx.doi.org/10.1182/blood-2006-05-...
).

Nonmitotic targets affecting the cell proliferation rate

Functional interphase proteins are targets that disrupt the cell cycle and prevent mitosis via DNA repair modulation, protein turnover and checkpoint signaling. Targeting these nonmitotic proteins may hinder cell division. For example, the ataxia-telangiectasia mutated (ATM) kinase induces a cell signaling cascade in response to DNA double-strand breaks. When activated, ATM phosphorylates substrates that promote DNA repair and, consequently, cell cycle progression (3838. Guo K, Shelat AA, Guy RK, Kastan MB. Development of a cell-based, high-throughput screening assay for ATM kinase inhibitors. J Biomol Screen. 2014;19(4):538-46, http://dx.doi.org/10.1177/1087057113520325.
http://dx.doi.org/10.1177/10870571135203...
). Somatic mutations in the ATM gene are found in several cancers, especially in hematological malignancies, and are often associated with chemotherapy resistance and unfavorable prognoses (3939. Lin K, Adamson J, Johnson GG, Carter A, Oates M, Wade R, et al. Functional analysis of the ATM-p53-p21 pathway in the LRF CLL4 trial: blockade at the level of p21 is associated with short response duration. Clin Cancer Res. 2012;18(15):4191-200, http://dx.doi.org/10.1158/1078-0432.CCR-11-2936.
http://dx.doi.org/10.1158/1078-0432.CCR-...
,4040. Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, et al. COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43(Database issue):D805-11, http://dx.doi.org/10.1093/nar/gku1075.
http://dx.doi.org/10.1093/nar/gku1075...
). Due to the substantial influence of the ATM kinase on cell cycle progression, the development of ATM-specific inhibitors is a good strategy in the search for new anticancer therapies (4141. Choi M, Kipps T, Kurzrock R. ATM Mutations in Cancer: therapeutic Implications. Mol Cancer Ther. 2016;15(8):1781-91, http://dx.doi.org/10.1158/1535-7163.MCT-15-0945.
http://dx.doi.org/10.1158/1535-7163.MCT-...
). For example, the ATM inhibitors CP466722 and Ku55933 were identified through a cell-based high-throughput screening assay (3838. Guo K, Shelat AA, Guy RK, Kastan MB. Development of a cell-based, high-throughput screening assay for ATM kinase inhibitors. J Biomol Screen. 2014;19(4):538-46, http://dx.doi.org/10.1177/1087057113520325.
http://dx.doi.org/10.1177/10870571135203...
).

WEE1 protein is a major tyrosine kinase involved in the progression of the cell cycle beyond the checkpoint between the G2 phase and mitosis (M phase), preventing entry into the mitotic cell division process in response to persistent DNA damage. High expression of this protein is often found in cancers (colorectal cancers, breast cancers, leukemias and other cancers) and correlates with tumor progression and negative prognoses, despite the cell cycle-arresting activity of WEE1 before mitosis onset (4242. Magnussen GI, Hellesylt E, Nesland JM, Trope CG, Flørenes VA, Holm R. High expression of wee1 is associated with malignancy in vulvar squamous cell carcinoma patients. BMC Cancer. 2013;13(1):288, http://dx.doi.org/10.1186/1471-2407-13-288.
http://dx.doi.org/10.1186/1471-2407-13-2...
43. Porter CC, Kim J, Fosmire S, Gearheart CM, van Linden A, Baturin D, et al. Integrated genomic analyses identify WEE1 as a critical mediator of cell fate and a novel therapeutic target in acute myeloid leukemia. Leukemia. 2012;26(6):1266-76, http://dx.doi.org/10.1038/leu.2011.392.
http://dx.doi.org/10.1038/leu.2011.392...
-4444. Slipicevic A, Holth A, Hellesylt E, Tropé CG, Davidson B, Flørenes VA. Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. Gynecol Oncol. 2014;135(1):118-24, http://dx.doi.org/10.1016/j.ygyno.2014.07.102.
http://dx.doi.org/10.1016/j.ygyno.2014.0...
). This apparent contradiction is not yet fully understood, and the current hypothesis is that WEE1 inhibition would promote the entry of cells with excessive DNA damage into mitosis, which would induce cell death during the ensuing M phase. WEE1 kinase inhibitors, e.g., AZD1775, have been studied in clinical trials, but their use is limited because of their nonselective action (4545. Wang Y, Li J, Booher RN, Kraker A, Lawrence T, Leopold WR, et al. Radiosensitization of p53 mutant cells by PD0166285, a novel G(2) checkpoint abrogator. Cancer Res. 2001;61(22):8211-7. 46. Palmer BD, Thompson AM, Booth RJ, Dobrusin EM, Kraker AJ, Lee HH, et al. 4-Phenylpyrrolo[3,4-c]carbazole-1,3(2H,6H)-dione inhibitors of the checkpoint kinase Wee1. Structure-activity relationships for chromophore modification and phenyl ring substitution. J Med Chem. 2006;49(16):4896-911, http://dx.doi.org/10.1021/jm0512591.
http://dx.doi.org/10.1021/jm0512591...
-4747. Matheson CJ, Backos DS, Reigan P. Targeting WEE1 Kinase in Cancer. Trends Pharmacol Sci. 2016;37(10):872-81, http://dx.doi.org/10.1016/j.tips.2016.06.006.
http://dx.doi.org/10.1016/j.tips.2016.06...
).

The 26S proteasome is an important multimeric complex responsible for the degradation of misfolded or overexpressed proteins (4848. Grigoreva TA, Tribulovich VG, Garabadzhiu AV, Melino G, Barlev NA. The 26S proteasome is a multifaceted target for anti-cancer therapies. Oncotarget. 2015;6(28):24733-49, http://dx.doi.org/10.18632/oncotarget.4619.
http://dx.doi.org/10.18632/oncotarget.46...
). Neoplastic cells exhibit high dependence on the 26S proteasome because of the need to eliminate defective proteins, which are detrimental to cell cycle advancement, resulting from the genetic instability in the tumor and the high cell proliferation rate. The accumulation of ubiquitinated and misfolded proteins may induce apoptosis. Therefore, the 26S proteasome is a validated therapeutic target in oncology, with FDA-approved inhibitors such as bortezomib (Velcade®) and carfilzomib currently in clinical use (4949. Moreau P. The emerging role of carfilzomib combination therapy in the management of multiple myeloma. Expert Rev Hematol. 2014;7(2):265-90, http://dx.doi.org/10.1586/17474086.2014.873699.
http://dx.doi.org/10.1586/17474086.2014....
) for the treatment of multiple myeloma, in which malignant cells have an abundance of proteasomes (4949. Moreau P. The emerging role of carfilzomib combination therapy in the management of multiple myeloma. Expert Rev Hematol. 2014;7(2):265-90, http://dx.doi.org/10.1586/17474086.2014.873699.
http://dx.doi.org/10.1586/17474086.2014....
,5050. Teicher BA, Tomaszewski JE, Tomaszewski JE. Proteasome inhibitors. Biochem Pharmacol. 2015;96(1):1-9, http://dx.doi.org/10.1016/j.bcp.2015.04.008.
http://dx.doi.org/10.1016/j.bcp.2015.04....
).

In addition to the proteins cited above, many others may be validated as targets for cancer therapy. A better understanding of mitotic mediators and their role in tumorigenesis may advance the fight against highly proliferative tumor cells.

Challenges in the search for new mitotic targets for anticancer therapy

Many efforts have been made in the search for and development of new antitumor compounds; however, cancer remains a difficult-to-treat disease (5151. Selvam C, Jordan BC, Prakash S, Mutisya D, Thilagavathi R. Pterocarpan scaffold: A natural lead molecule with diverse pharmacological properties. Eur J Med Chem. 2017;128:219-36, http://dx.doi.org/10.1016/j.ejmech.2017.01.023.
http://dx.doi.org/10.1016/j.ejmech.2017....
). The available antimitotic therapies are somewhat successful but rarely eradicate tumors completely and generally promote tumor resistance and systemic toxicity (5252. Chan KS, Koh CG, Li HY. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis. 2012;3(10):e411, http://dx.doi.org/10.1038/cddis.2012.148.
http://dx.doi.org/10.1038/cddis.2012.148...
). The selectivity of antimitotics for tumors has long been believed to be based on the high proliferation rate of cancer cells in contrast to the supposed quiescence or low division rate of most cells in normal tissues. Normal and tumor cells progress through the cell cycle in similar ways, but the higher proliferation rates of cancer cells cause mitotic targets to be more frequently expressed in tumors than in normal tissues, thus making neoplastic cells more sensitive to antimitotic action (5252. Chan KS, Koh CG, Li HY. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis. 2012;3(10):e411, http://dx.doi.org/10.1038/cddis.2012.148.
http://dx.doi.org/10.1038/cddis.2012.148...
). However, Komlodi-Pasztor and colleagues emphasized that tumor proliferation rates may not be as high as expected, and thus the frequency of mitotic target expression may not be as different as expected between tumor and nontumor cell populations. This characteristic could be a reason for the lack of specificity of antimitotic agents such as mitosis-specific kinase inhibitors (5353. Komlodi-Pasztor E, Sackett DL, Fojo AT. Inhibitors targeting mitosis: tales of how great drugs against a promising target were brought down by a flawed rationale. Clin Cancer Res. 2012;18(1):51-63, http://dx.doi.org/10.1158/1078-0432.CCR-11-0999.
http://dx.doi.org/10.1158/1078-0432.CCR-...
).

In addition to selectivity issues, antimitotic drugs acting only on M phase-exclusive targets allow interphase cancer cells to be refractory to treatment. In his review, Mitchison (5454. Mitchison TJ. The proliferation rate paradox in antimitotic chemotherapy. Mol Biol Cell. 2012;23(1):1-6, http://dx.doi.org/10.1091/mbc.e10-04-0335.
http://dx.doi.org/10.1091/mbc.e10-04-033...
) compared data from the literature and showed that 2-5% of cells in primary tumors are in the S phase, while 40% of cells in in vitro tumor cell cultures are in the S phase. In addition, the doubling times of tumor cells in patients are very long, approximately fifty times greater than those in cell lines or animal models. Furthermore, the cell proliferation ratio varies greatly according to different cancer types, individual patient characteristics, and the primary tumor origin and location. Therefore, the number of neoplastic cells in interphase is probably high enough to make a tumor resistant to chemotherapeutics that act specifically on exclusive mitotic targets. This fact may explain the preclinical failure of many antimitotic compounds (5252. Chan KS, Koh CG, Li HY. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis. 2012;3(10):e411, http://dx.doi.org/10.1038/cddis.2012.148.
http://dx.doi.org/10.1038/cddis.2012.148...
,5555. Manchado E, Guillamot M, Malumbres M. Killing cells by targeting mitosis. Cell Death Differ. 2012;19(3):369-77, http://dx.doi.org/10.1038/cdd.2011.197.
http://dx.doi.org/10.1038/cdd.2011.197...
).

Antimitotics are considered mitosis blockers and, consequently, cell death inducers. The main issue in antimitotic therapy is that not all cells blocked in M phase will die, with many becoming aneuploid. This alteration is a direct consequence of chromosomal instability, that is, susceptibility to the gain and/or loss of chromosomes, which generates an unstable karyotype. This instability promotes the acquisition of malignant tumor characteristics as a proliferative advantage due to the increased mutation rate in oncogenes and tumor suppressor genes (5555. Manchado E, Guillamot M, Malumbres M. Killing cells by targeting mitosis. Cell Death Differ. 2012;19(3):369-77, http://dx.doi.org/10.1038/cdd.2011.197.
http://dx.doi.org/10.1038/cdd.2011.197...
). The gain of malignant features by tumors hinders the prediction of patient response to therapy and facilitates the emergence of chemotherapy-resistant tumor cells. Treatment resistance is a major problem that hinders complete cure and limits the success of anticancer drug therapy. For example, the differential expression of β-tubulin isoforms has been shown to result in resistance to anti-tubulin chemotherapeutics such as paclitaxel, along with increasing tumor aggressiveness (5656. Freedman H, Huzil JT, Luchko T, Ludueãa RF, Tuszynski JA. Identification and characterization of an intermediate taxol binding site within microtubule nanopores and a mechanism for tubulin isotype binding selectivity. J Chem Inf Model. 2009;49(2):424-36, http://dx.doi.org/10.1021/ci8003336.
http://dx.doi.org/10.1021/ci8003336...
,5757. Ferrandina G, Zannoni GF, Martinelli E, Paglia A, Gallotta V, Mozzetti S, et al. Class III β-tubulin overexpression is a marker of poor clinical outcome in advanced ovarian cancer patients. Clin Cancer Res. 2006;12(9):2774-9, http://dx.doi.org/10.1158/1078-0432.CCR-05-2715.
http://dx.doi.org/10.1158/1078-0432.CCR-...
). Another emerging form of taxane resistance is the increased expression of multidrug resitance (MDR) proteins – efflux pumps that remove cancer drugs from the tumor cell cytoplasm – such as the ATP Binding Cassette Subfamily B Member 1 (ABCB1 protein), P-glycoprotein and the Multidrug resistance-associated protein 1 (MRP1) (5858. Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist. 2003;8(5):411-24, http://dx.doi.org/10.1634/theoncologist.8-5-411.
http://dx.doi.org/10.1634/theoncologist....
,5959. Fojo AT, Menefee M. Microtubule targeting agents: basic mechanisms of multidrug resistance (MDR). Semin Oncol. 2005;32(6 Suppl 7):S3-8, http://dx.doi.org/10.1053/j.seminoncol.2005.09.010.
http://dx.doi.org/10.1053/j.seminoncol.2...
).

The side effects of antimitotics are also related to the ubiquitous function of targets such as microtubules, which act in mitosis, interphase and quiescence. Drugs that disrupt microtubule function are associated with side effects such as neurotoxicity, which may lead to irreversible neuropathies. The current generation of antimitotic therapies in clinical trials did not meet the expectations of reduced side effects, even with diminished neurotoxicity (6060. Doménech E, Malumbres M. Mitosis-targeting therapies: a troubleshooting guide. Curr Opin Pharmacol. 2013;13(4):519-28, http://dx.doi.org/10.1016/j.coph.2013.03.011.
http://dx.doi.org/10.1016/j.coph.2013.03...
). In fact, new side effects were found in patients treated with antimitotics and included hematological changes such as neutropenia (6161. Schöffski P, Awada A, Dumez H, Gil T, Bartholomeus S, Wolter P, et al. A phase I, dose-escalation study of the novel Polo-like kinase inhibitor volasertib (BI 6727) in patients with advanced solid tumours. Eur J Cancer. 2012;48(2):179-86, http://dx.doi.org/10.1016/j.ejca.2011.11.001.
http://dx.doi.org/10.1016/j.ejca.2011.11...
62. Arkenau HT, Plummer R, Molife LR, Olmos D, Yap TA, Squires M, et al. A phase I dose escalation study of AT9283, a small molecule inhibitor of aurora kinases, in patients with advanced solid malignancies. Ann Oncol. 2012;23(5):1307-13, http://dx.doi.org/10.1093/annonc/mdr451.
http://dx.doi.org/10.1093/annonc/mdr451...
-6363. Jones R, Vuky J, Elliott T, Mead G, Arranz JA, Chester J, et al. Phase II study to assess the efficacy, safety and tolerability of the mitotic spindle kinesin inhibitor AZD4877 in patients with recurrent advanced urothelial cancer. Invest New Drugs. 2013;31(4):1001-7, http://dx.doi.org/10.1007/s10637-013-9926-y.
http://dx.doi.org/10.1007/s10637-013-992...
). Next, we review the novel classes of antimitotics under preclinical investigation by our research group, which have the potential to overcome or minimize the limitations of current anticancer antimitotics. These molecules are synthetic analogs of well-known microtubule-targeting agents (combretastatins) and natural products with multiple or unknown targets (withanolides and pterocarpans).

Combretastatins

The combretastatins are a potent class of natural products first isolated by Pettit and collaborators from the bark of the African bush willow, Combretum caffrum (6464. Fürst R, Zupkó I, Berényi A, Ecker GF, Rinner U. Synthesis and antitumor-evaluation of cyclopropyl-containing combretastatin analogs. Bioorg Med Chem Lett. 2009;19(24):6948-51, http://dx.doi.org/10.1016/j.bmcl.2009.10.064.
http://dx.doi.org/10.1016/j.bmcl.2009.10...
,6565. Shan Y, Zhang J, Liu Z, Wang M, Dong Y. Developments of combretastatin A-4 derivatives as anticancer agents. Curr Med Chem. 2011;18(4):523-38, http://dx.doi.org/10.2174/092986711794480221.
http://dx.doi.org/10.2174/09298671179448...
). These molecules are phenolic stilbenes that bind to the colchicine binding site on tubulin and lead to microtubule disruption and mitotic arrest by preventing spindle formation (6666. Seddigi ZS, Malik MS, Saraswati AP, Ahmed SA, Babalghith AO, Lamfon HA, et al. Recent advances in combretastatin based derivatives and prodrugs as antimitotic agents. MedChemComm. 2017;8(8):1592-603, http://dx.doi.org/10.1039/C7MD00227K.
http://dx.doi.org/10.1039/C7MD00227K...
). Among the combretastatins, combretastatin A-4 (CA-4) is a lead compound that binds to the colchicine site and exerts antiproliferative activity by inhibiting tubulin polymerization. Despite its structural simplicity and potent activity, this natural product has problems related to its low solubility in natural biological systems and the lability of the cis-stilbene moiety. However, CA-4 is one of the most actively researched lead molecules in anticancer drug development programs worldwide, and extensive studies have been conducted to develop new CA-4 analogs to overcome the issues inherent to CA-4 and improve its pharmacotherapeutic profile (6767. Yang T, Wang Y, Li Z, Dai W, Yin J, Liang L, et al. Targeted delivery of a combination therapy consisting of combretastatin A4 and low-dose doxorubicin against tumor neovasculature. Nanomedicine (Lond). 2012;8(1):81-92, http://dx.doi.org/10.1016/j.nano.2011.05.003.
http://dx.doi.org/10.1016/j.nano.2011.05...
).

Most of the recent structural modifications introduced in CA-4 have been based on changes in the cis olefinic bond, including the restriction of the cis configuration by a heterocyclic moiety and the replacement of this configuration by a stable linker. These modifications can maintain the original aromatic rings or can be associated with simultaneous alterations in either ring A or ring B (6868. Combes S, Barbier P, Douillard S, McLeer-Florin A, Bourgarel-Rey V, Pierson JT, et al. Synthesis and biological evaluation of 4-arylcoumarin analogues of combretastatins. Part 2. J Med Chem. 2011;54(9):3153-62, http://dx.doi.org/10.1021/jm901826e.
http://dx.doi.org/10.1021/jm901826e...
). Among the new linkers explored to replace the cis olefinic bond of the CA-4 structure, N-acylhydrazone (NAH) is worth mentioning. From the synthetic and medicinal chemistry perspectives, NAH is considered a versatile subunit, and it has been described as a privileged structure (6969. Duarte CD, Barreiro EJ, Fraga CA. Privileged structures: a useful concept for the rational design of new lead drug candidates. Mini Rev Med Chem. 2007;7(11):1108-19, http://dx.doi.org/10.2174/138955707782331722.
http://dx.doi.org/10.2174/13895570778233...
). This review highlights some NAH derivatives that perturb microtubule/tubulin dynamics and have been synthesized by our research group using LASSBio® compounds from the Federal University of Rio de Janeiro.

Amaral et al. (7070. do Amaral DN, Cavalcanti BC, Bezerra DP, Ferreira PM, Castro RP, Sabino JR, et al. Docking, synthesis and antiproliferative activity of N-acylhydrazone derivatives designed as combretastatin A4 analogues. PLoS One. 2014;9(3):e85380, http://dx.doi.org/10.1371/journal.pone.0085380.
http://dx.doi.org/10.1371/journal.pone.0...
) identified a new series of CA-4 analogs designed in light of previous structure–activity relationship studies on CA-4 derivatives (7171. Hu L, Li ZR, Li Y, Qu J, Ling YH, Jiang JD, et al. Synthesis and structure-activity relationships of carbazole sulfonamides as a novel class of antimitotic agents against solid tumors. J Med Chem. 2006;49(21):6273-82, http://dx.doi.org/10.1021/jm060546h.
http://dx.doi.org/10.1021/jm060546h...
). Several compounds exhibit moderate to high antiproliferative activity in tumor cells, with IC50 values ranging between 18 and 4 nmol.L-1. Among these compounds, LASSBio-1586, patent number WO2013142935A1, has emerged as a lead antitumor candidate; LASSBio-1586 is structurally simple, able to inhibit microtubule polymerization, exhibits a broad in vitro and in vivo antiproliferative profile and has a better cytotoxic selectivity index than the natural prototype CA-4 (7070. do Amaral DN, Cavalcanti BC, Bezerra DP, Ferreira PM, Castro RP, Sabino JR, et al. Docking, synthesis and antiproliferative activity of N-acylhydrazone derivatives designed as combretastatin A4 analogues. PLoS One. 2014;9(3):e85380, http://dx.doi.org/10.1371/journal.pone.0085380.
http://dx.doi.org/10.1371/journal.pone.0...
). In addition, the antiproliferative activity was evaluated in human lymphocytes to determine the selectivity indexes of the LASSBio derivatives, which were better than that of CA-4 (7272. Cavalcanti BC, Barros FW, Cabral IO, Ferreira JR, Magalhães HI, Júnior HV, et al. Preclinical genotoxicology of nor-β-lapachone in human cultured lymphocytes and Chinese hamster lung fibroblasts. Chem Res Toxicol. 2011;24(9):1560-74, http://dx.doi.org/10.1021/tx200180y.
http://dx.doi.org/10.1021/tx200180y...
). More recently, our group described the synthesis of nine new isosteres of LASSBio-1586, aimed at improving the cytotoxicity and selectivity of LASSBio-1586 (7373. Carneiro TR, Amaral DN, Fokoue HH, Sant’Anna CM, Porras ML, Oliveira AC. Synthesis, pharmacological evaluation and docking study of a new modulator of microtubule polymerization. Lett Drug Design Disc. 2018;15(7):778-86, http://dx.doi.org/10.2174/1570180814666171012162557.
http://dx.doi.org/10.2174/15701808146661...
). Among these derivatives, LASSBio-1920 was selected as the most potent, with IC50 values of 0.3, 9.0 and 5.0 nmol.L-1 in the HL-60, OVCAR-8 and HCT-8 tumor cell lines, respectively (Table 1). LASSBio-1920 also exhibited a notable cytotoxic selectivity index in comparison with that of CA-4 and LASSBio-1586 (Table 2, Figure 2), and its ability to modulate microtubule polymerization was confirmed. Docking studies indicated that LASSBio-1920 can interact with the same amino acid residues involved in the interaction of DAMA-colchicine as a cocrystal with β-tubulin.

Table 1
Cytotoxic activity (mean IC50±SEM) of LASSBio-1586 analogs and the CA-4 and LASSBio-1586 standards in tumor cell lines and peripheral blood mononuclear cells (PBMCs) after 72h of treatment (IC50 values are in µmol.L-1). SEM, standard error of the mean; IC50, the concentration of a compound needed to reduce cell growth by 50% in vitro.

Table 2
Cytotoxic selectivity index (SI), which represents the IC50 for PBMCs/cancer cell lines after 72h of treatment. PBMCs, peripheral blood mononuclear cells; SEM, standard error of the mean; IC50, the concentration of a compound needed to reduce cell growth by 50% in vitro.

Figure 2
Combretastatin A-4 analogs. (A) LASSBio-1586. (B) LASSBio-1920.

Our data from structure–activity relationship studies on combretastatins reveal that the presence of the methoxy (OCH3) group linked at position 5 of the aromatic subunit (ring A) is crucial for cytotoxic activity, while the OCH3 group at position 4 is not. Concerning the aromatic rings linked to the imine subunit (ring B) of the isostere, our data suggest a direct relationship between the increase in lipophilicity and the enhancement of cytotoxic activity, in agreement with previous results of Amaral et al. (7070. do Amaral DN, Cavalcanti BC, Bezerra DP, Ferreira PM, Castro RP, Sabino JR, et al. Docking, synthesis and antiproliferative activity of N-acylhydrazone derivatives designed as combretastatin A4 analogues. PLoS One. 2014;9(3):e85380, http://dx.doi.org/10.1371/journal.pone.0085380.
http://dx.doi.org/10.1371/journal.pone.0...
). These findings can be explained on the basis of the improved hydrophobic interaction with β-tubulin (7070. do Amaral DN, Cavalcanti BC, Bezerra DP, Ferreira PM, Castro RP, Sabino JR, et al. Docking, synthesis and antiproliferative activity of N-acylhydrazone derivatives designed as combretastatin A4 analogues. PLoS One. 2014;9(3):e85380, http://dx.doi.org/10.1371/journal.pone.0085380.
http://dx.doi.org/10.1371/journal.pone.0...
). The more lipophilic the group bound to ring B is, the stronger the interaction with β-tubulin. However, the addition of lipophilic/hydrophobic groups to the combretastatin core should be carefully considered, since this structural modification may impair aqueous solubility and pharmacokinetic properties.

On the basis of the articles discussed here, we observed that microtubules and their associated proteins are feasible targets in drug discovery programs, particularly for the treatment of cancer. Numerous potent CA-4 derivatives have been developed in recent years, although very few have been accepted for clinical studies. Therefore, further structure-based drug discovery and optimization steps focusing on the CA-4 molecule and its analogs should be supported and encouraged to overcome the limitations of CA-4.

Withanolides

Withanolides are lactones based on the cholesterol-derived ergostane molecule, with oxygen atoms enzymatically added to carbons of the steroidal core and its side chains, thus producing complex and unusual structures. These compounds are secondary metabolites found in plants of the family Solanaceae (7474. Glotter E. Withanolides and related ergostane-type steroids. Nat Prod Rep. 1991;8(4):415-40, http://dx.doi.org/10.1039/np9910800415.
http://dx.doi.org/10.1039/np9910800415...
) and exhibit multiple biological activities, such as antitumor (7575. Zhu F, Dai C, Fu Y, Loo JF, Xia D, Gao SP, et al. Physalin A exerts anti-tumor activity in non-small cell lung cancer cell lines by suppressing JAK/STAT3 signaling. Oncotarget. 2016;7(8):9462-76, http://dx.doi.org/10.18632/oncotarget.7051.
http://dx.doi.org/10.18632/oncotarget.70...
), cytotoxic (7676. Rocha DD, Militão GC, Veras ML, Pessoa OD, Silveira ER, Alves AP, et al. Selective cytotoxicity of withaphysalins in myeloid leukemia cell lines versus peripheral blood mononuclear cells. Life Sci. 2006;79(18):1692-701, http://dx.doi.org/10.1016/j.lfs.2006.05.026.
http://dx.doi.org/10.1016/j.lfs.2006.05....
), antistress (7777. Kour K, Pandey A, Suri KA, Satti NK, Gupta KK, Bani S. Restoration of stress-induced altered T cell function and corresponding cytokines patterns by Withanolide A. Int Immunopharmacol. 2009;9(10):1137-44, http://dx.doi.org/10.1016/j.intimp.2009.05.011.
http://dx.doi.org/10.1016/j.intimp.2009....
), antifeedant (7878. Mareggiani G, Picollo MI, Zerba E, Burton G, Tettamanzi MC, Benedetti-Doctorovich MO, et al. Antifeedant activity of withanolides from Salpichroa origanifolia on Musca domestica. J Nat Prod. 2000;63(8):1113-6, http://dx.doi.org/10.1021/np0001068.
http://dx.doi.org/10.1021/np0001068...
), immunosuppressive (7979. Soares MB, Brustolim D, Santos LA, Bellintani MC, Paiva FP, Ribeiro YM, et al. Physalins B, F and G, seco-steroids purified from Physalis angulata L., inhibit lymphocyte function and allogeneic transplant rejection. Int Immunopharmacol. 2006;6(3):408-14, http://dx.doi.org/10.1016/j.intimp.2005.09.007.
http://dx.doi.org/10.1016/j.intimp.2005....
) and antimicrobial (8080. Shanazbanu S, Shashidara S, Babu VL, Dhanapal R. Isolation of withaferin A from Withania somnifera Dun leaves and its antibacterial activity. Asian J Chem. 2006;18(2):1243-7.) activities.

One of the most heavily investigated withanolides is withaferin A, which was originally isolated from the medicinal plant Withania somnifera (L.) Dunal, also called Ashwagandha or Indian ginseng. This molecule exhibits growth-inhibitory activity in many tumor cell lines through multiple mechanisms, including an antimitotic mode of action. Like taxanes and vinca alkaloids, withaferin A binds to β-tubulin, disorganizes the mitotic spindle and causes cell cycle arrest at the G2/M phase transition in breast cancer cells (8181. Antony ML, Lee J, Hahm ER, Kim SH, Marcus AI, Kumari V, et al. Growth arrest by the antitumor steroidal lactone withaferin A in human breast cancer cells is associated with down-regulation and covalent binding at cysteine 303 of β-tubulin. J Biol Chem. 2014;289(3):1852-65, http://dx.doi.org/10.1074/jbc.M113.496844.
http://dx.doi.org/10.1074/jbc.M113.49684...
). This compound also impairs spindle assembly checkpoint function (8282. Das T, Roy KS, Chakrabarti T, Mukhopadhyay S, Roychoudhury S. Withaferin A modulates the Spindle assembly checkpoint by degradation of Mad2-Cdc20 complex in colorectal cancer cell lines. Biochem Pharmacol. 2014;91(1):31-9, http://dx.doi.org/10.1016/j.bcp.2014.06.022.
http://dx.doi.org/10.1016/j.bcp.2014.06....
) and alters the expression levels of mitosis-related proteins, such as WEE1, phospho-histone H3, cyclin-dependent kinase inhibitor 1 (p21 protein), Aurora B (8383. Roy RV, Suman S, Das TP, Luevano JE, Damodaran C. Withaferin-A induces mitotic catastrophe and growth arrest in prostate cancer cells. J Nat Prod. 2013;76(10):1909-15, http://dx.doi.org/10.1021/np400441f.
http://dx.doi.org/10.1021/np400441f...
) and the anaphase-promoting complex (APC) substrate securin (8484. Stan SD, Zeng Y, Singh SV. Ayurvedic medicine constituent withaferin a causes G2 and M phase cell cycle arrest in human breast cancer cells. Nutr Cancer. 2008;60(Suppl 1):51-60, http://dx.doi.org/10.1080/01635580802381477.
http://dx.doi.org/10.1080/01635580802381...
). Withaferin A also targets a variety of nonmitotic proteins, such as vimentin (8585. Bargagna-Mohan P, Hamza A, Kim YE, Khuan Abby Ho Y, Mor-Vaknin N, Wendschlag N, et al. The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. Chem Biol. 2007;14(6):623-34, http://dx.doi.org/10.1016/j.chembiol.2007.04.010.
http://dx.doi.org/10.1016/j.chembiol.200...
), annexin II (8686. Falsey RR, Marron MT, Gunaherath GM, Shirahatti N, Mahadevan D, Gunatilaka AA, et al. Actin microfilament aggregation induced by withaferin A is mediated by annexin II. Nat Chem Biol. 2006;2(1):33-8, http://dx.doi.org/10.1038/nchembio755.
http://dx.doi.org/10.1038/nchembio755...
), heat shock protein 90 (8787. Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B, et al. Withaferin A targets heat shock protein 90 in pancreatic cancer cells. Biochem Pharmacol. 2010;79(4):542-51, http://dx.doi.org/10.1016/j.bcp.2009.09.017.
http://dx.doi.org/10.1016/j.bcp.2009.09....
) and the proteasome (8888. Yang H, Shi G, Dou QP. The tumor proteasome is a primary target for the natural anticancer compound Withaferin A isolated from “Indian winter cherry”. Mol Pharmacol. 2007;71(2):426-37, http://dx.doi.org/10.1124/mol.106.030015.
http://dx.doi.org/10.1124/mol.106.030015...
). Although these proteins function mainly during interphase, affecting them may prevent mitosis and inhibit cell cycle progression. The exact mechanism of action of withaferin A is not fully understood and may encompass different cell functions during different cell cycle phases.

Another group of withanolides called the withaphysalins was isolated from the plant Acnistus arborescens. Members of this group have chemical structures closely related to those of withaferin A and have also been studied by our research team (8989. Veras ML, Bezerra MZ, Braz-Filho R, Pessoa OD, Montenegro RC, do O Pessoa C, et al. Cytotoxic epimeric withaphysalins from leaves of Acnistus arborescens. Planta Med. 2004;70(6):551-5, http://dx.doi.org/10.1055/s-2004-827156.
http://dx.doi.org/10.1055/s-2004-827156...
90. Veras ML, Bezerra MZ, Lemos TL, Uchoa DE, Braz-Filho R, Chai HB, et al. Cytotoxic withaphysalins from the leaves of Acnistus arborescens. J Nat Prod. 2004;67(4):710-3, http://dx.doi.org/10.1021/np0340347.
http://dx.doi.org/10.1021/np0340347...
-9191. Rocha DD, Balgi A, Maia AI, Pessoa OD, Silveira ER, Costa-Lotufo LV, et al. Cell cycle arrest through inhibition of tubulin polymerization by withaphysalin F, a bioactive compound isolated from Acnistus arborescens. Invest New Drugs. 2012;30(3):959-66, http://dx.doi.org/10.1007/s10637-011-9649-x.
http://dx.doi.org/10.1007/s10637-011-964...
). Rocha et al. showed that withaphysalin F and withaferin A may have mechanisms in common because the antiproliferative activity of withaphysalin F and its effect of arresting cells at the G2/M phase transition were also due to interference with microtubule polymerization (9191. Rocha DD, Balgi A, Maia AI, Pessoa OD, Silveira ER, Costa-Lotufo LV, et al. Cell cycle arrest through inhibition of tubulin polymerization by withaphysalin F, a bioactive compound isolated from Acnistus arborescens. Invest New Drugs. 2012;30(3):959-66, http://dx.doi.org/10.1007/s10637-011-9649-x.
http://dx.doi.org/10.1007/s10637-011-964...
).

Pterocarpans

Pterocarpans are compounds extracted from the plant families Fabaceae, Leguminosae, Papilionaceae and Bituminaria and comprise the second largest group of isoflavonoids. Pterocarpans are phytoalexins produced by plants to defend against adversities such as resource shortages, pathogens and UV radiation (9292. Aoki T, Akashi T, Ayabe S. Flavonoids of leguminous plants: structure, biological activity, and biosynthesis. J Plant Res. 2000;113(4):475-88, http://dx.doi.org/10.1007/PL00013958.
http://dx.doi.org/10.1007/PL00013958...
). In ethnopharmacology, these compounds are extensively used to treat different ailments. These compounds have interesting biological activities, such as snake antivenom (9393. Nakagawa M, Nakanishi K, Darko LL, Vick JA. Structures of cabenegrins AI and A-II, potent anti-snake venoms. Tetrahedron Lett. 1982;23(38):3855-8, http://dx.doi.org/10.1016/S0040-4039(00)87726-6.
http://dx.doi.org/10.1016/S0040-4039(00)...
), anti-HIV (9494. Engler TA, Lynch KO, Reddy JP, Gregory GS. Synthetic pterocarpans with anti-HIV activity. Bioorg Med Chem Lett. 1993;3(6):1229-32, http://dx.doi.org/10.1016/S0960-894X(00)80321-2.
http://dx.doi.org/10.1016/S0960-894X(00)...
), anti-Trypanosoma spp., anti-Leishmania spp. (9595. Salem MM, Werbovetz KA. Isoflavonoids and other compounds from Psorothamnus arborescens with antiprotozoal activities. J Nat Prod. 2006;69(1):43-9, http://dx.doi.org/10.1021/np0502600.
http://dx.doi.org/10.1021/np0502600...
), antifungal, antibiotic (9696. Jiménez-González L, Álvarez-Corral M, Muãoz-Dorado M, Rodríguez-García I. Pterocarpans: interesting natural products with antifungal activity and other biological properties. Phytochem Rev. 2008;7(1):125-54.), and topoisomerase I-inhibiting activities (9797. Tesauro C, Fiorani P, D’Annessa I, Chillemi G, Turchi G, Desideri A, Erybraedin C. A natural compound from the plant Bituminaria bituminosa, inhibits both the cleavage and religation activities of human topoisomerase I. Biochem J. 2010;425(3):531-9, http://dx.doi.org/10.1042/BJ20091127.
http://dx.doi.org/10.1042/BJ20091127...
). Pterocarpans have a benzofuran–benzopyran tetracyclic ring system, which contains two chiral centers derived from the flavonoid skeleton at the 6a and 11a positions (9696. Jiménez-González L, Álvarez-Corral M, Muãoz-Dorado M, Rodríguez-García I. Pterocarpans: interesting natural products with antifungal activity and other biological properties. Phytochem Rev. 2008;7(1):125-54.,9898. Goel A, Kumar A, Raghuvanshi A. Synthesis, stereochemistry, structural classification, and chemical reactivity of natural pterocarpans. Chem Rev. 2013;113(3):1614-40, http://dx.doi.org/10.1021/cr300219y.
http://dx.doi.org/10.1021/cr300219y...
).

Hundreds of different pterocarpans have been discovered through different extraction methods, and the pharmacological activity of these pterocarpans has been tested. Since the basic structure of these compounds is chiral, methods to synthesize pterocarpans have been suggested, for example, sodium borohydride reduction of 2’-hydroxy isoflavones, Heck arylation of 2H-chromenes, Claisen rearrangement of aryl allyl ethers, aldol condensation between phenylacetates and benzaldehydes, 5-endo-trig radical cyclization, alkene metathesis and many others (9898. Goel A, Kumar A, Raghuvanshi A. Synthesis, stereochemistry, structural classification, and chemical reactivity of natural pterocarpans. Chem Rev. 2013;113(3):1614-40, http://dx.doi.org/10.1021/cr300219y.
http://dx.doi.org/10.1021/cr300219y...
). (+)2,3,9-Trimethoxypterocarpan was isolated from Platymiscium floribundum in Brazil, and the convergent syntheses of its racemic and enantiomerically pure forms was developed by Dr. M. Banwell at the Australian National University (ANU). The synthetic route was patented in 2012 [WO2013000054 (A1)]. The molecule synthesized by Professor Banwell showed the same 1H NMR spectrum as the natural product (9999. Falcão MJ, Pouliquem YB, Lima MA, Gramosa NV, Costa-Lotufo LV, Militão GC, et al. Cytotoxic flavonoids from Platymiscium floribundum. J Nat Prod. 2005;68(3):423-6, http://dx.doi.org/10.1021/np049854d.
http://dx.doi.org/10.1021/np049854d...
).

The compound (+)2,3,9-trimethoxypterocarpan has been investigated by our research group (Figure 3) as a potential anticancer compound (9999. Falcão MJ, Pouliquem YB, Lima MA, Gramosa NV, Costa-Lotufo LV, Militão GC, et al. Cytotoxic flavonoids from Platymiscium floribundum. J Nat Prod. 2005;68(3):423-6, http://dx.doi.org/10.1021/np049854d.
http://dx.doi.org/10.1021/np049854d...
). This molecule showed cytotoxicity against four leukemia cell lines (HL-60, Molt-4, Jurkat, and K562), and the proliferation of K562 cells was blocked after a 48-hour incubation period (at an IC50 of 0.8 µg.mL-1) (9999. Falcão MJ, Pouliquem YB, Lima MA, Gramosa NV, Costa-Lotufo LV, Militão GC, et al. Cytotoxic flavonoids from Platymiscium floribundum. J Nat Prod. 2005;68(3):423-6, http://dx.doi.org/10.1021/np049854d.
http://dx.doi.org/10.1021/np049854d...
,100100. Militão GC, Bezerra DP, Pessoa C, de Moraes MO, da Ponte FA, Lima MA, et al. Comparative cytotoxicity of 2, 3, 9-trimethoxypterocarpan in leukemia cell lines (HL-60, Jurkat, Molt-4, and K562) and human peripheral blood mononuclear cells. J Nat Med. 2007;61(2):196-9, http://dx.doi.org/10.1007/s11418-006-0118-3.
http://dx.doi.org/10.1007/s11418-006-011...
). Flow cytometric analysis of HL-60 cells treated for 3 hours with (+)2,3,9-trimethoxypterocarpan showed the inhibition of DNA synthesis using a bromodeoxyuridine (BrdU) uptake assay. The pterocarpan inhibited BrdU incorporation by 37, 38 and 56% during the S phase at concentrations of 3.97, 7.95 and 15.9 μmol.L-1, respectively. After a 24-hour treatment with the same concentrations, propidium iodide-stained HL-60 cells showed no significant membrane damage but DNA fragmentation was seen in almost 50% of cells permeabilized with Triton X-100. Most HL-60 cells were arrested at the G2/M phase transition with tetraploid DNA content after treatment with 15.9 μmol.L-1 (+)2,3,9-trimethoxypterocarpan. In the indirect determination of mitochondrial depolarization via rhodamine 123, 38.6% of the HL-60 cell population exhibited depolarized mitochondria at the lowest treatment concentration. The results of the BrdU, membrane integrity and DNA content experiments suggested that (+)2,3,9-trimethoxypterocarpan is a cytostatic agent, while the results of the DNA fragmentation and mitochondrial depolarization experiments suggested that this pterocarpan induces apoptosis in HL-60 cells (101101. Militão GC, Dantas IN, Pessoa C, Falcão MJ, Silveira ER, Lima MA, et al. Induction of apoptosis by pterocarpans from Platymiscium floribundum in HL-60 human leukemia cells. Life Sci. 2006;78(20):2409-17, http://dx.doi.org/10.1016/j.lfs.2005.09.044.
http://dx.doi.org/10.1016/j.lfs.2005.09....
).

Figure 3
Structure of pterocarpans isolated from Platymiscium floribundum by Falcão et al. (9595. Salem MM, Werbovetz KA. Isoflavonoids and other compounds from Psorothamnus arborescens with antiprotozoal activities. J Nat Prod. 2006;69(1):43-9, http://dx.doi.org/10.1021/np0502600.
http://dx.doi.org/10.1021/np0502600...
).

The cytostatic activity of (+)2,3,9-trimethoxypterocarpan in leukemia cells was corroborated by immunocytochemical experiments in the human breast adenocarcinoma cell lines MCF-7, T47D and HS58T. After treatment with 7.95 μmol.L-1 (+)2,3,9-trimethoxypterocarpan, the breast cancer cells remained arrested in mitosis with intact actin and microtubule networks. The same result was observed in nontumor BRL3A (murine liver) cells. However, the arrangement of microtubules was modified in most dividing cells, which exhibited monopolar mitotic spindles. Tripolar spindles were also observed, mainly in T47D cells (Figure 4). Such aberrant configurations prevented the alignment of chromosomes on the metaphase plate, blocking cell cycle progression and, consequently, cell division. Interestingly, none of the treated interphase cells exhibited microtubules with morphological alterations, suggesting that the effect of (+)2,3,9-trimethoxypterocarpan is restricted to mitosis. Lamin B labeling showed nuclear envelope disruption and confirmed cell cycle arrest at prometaphase (102102. Militão GC, Prado MP, Pessoa C, de Moraes MO, Silveira ER, Lima MA, et al. Pterocarpans induce tumor cell death through persistent mitotic arrest during prometaphase. Biochimie. 2014;104:147-55, http://dx.doi.org/10.1016/j.biochi.2014.06.005.
http://dx.doi.org/10.1016/j.biochi.2014....
). The monastral spindles observed in treated breast cancer cells resembled the phenotype induced by monastrol treatment (103103. Klein E, DeBonis S, Thiede B, Skoufias DA, Kozielski F, Lebeau L. New chemical tools for investigating human mitotic kinesin Eg5. Bioorg Med Chem. 2007;15(19):6474-88, http://dx.doi.org/10.1016/j.bmc.2007.06.016.
http://dx.doi.org/10.1016/j.bmc.2007.06....
,104104. Müller C, Gross D, Sarli V, Gartner M, Giannis A, Bernhardt G, et al. Inhibitors of kinesin Eg5: antiproliferative activity of monastrol analogues against human glioblastoma cells. Cancer Chemother Pharmacol. 2007;59(2):157-64.). Monastrol impairs the acquisition of bipolarity in dividing cells without affecting centriole duplication (105105. Kapoor TM, Mayer TU, Coughlin ML, Mitchison TJ. Probing spindle assembly mechanisms with monastrol, a small molecule inhibitor of the mitotic kinesin, Eg5. J Cell Biol. 2000;150(5):975-88, http://dx.doi.org/10.1083/jcb.150.5.975.
http://dx.doi.org/10.1083/jcb.150.5.975...
), apparently due to its activity against the Eg5 kinesin (106106. Gartner M, Sunder-Plassmann N, Seiler J, Utz M, Vernos I, Surrey T, et al. Development and biological evaluation of potent and specific inhibitors of mitotic Kinesin Eg5. ChemBioChem. 2005;6(7):1173-7, http://dx.doi.org/10.1002/cbic.200500005.
http://dx.doi.org/10.1002/cbic.200500005...
).

Figure 4
Formation of atypical mitotic spindles in 2,3,9-trimethoxypterocarpan-treated cells. Actin (green), tubulin (blue) and nuclei (red) are labeled in breast carcinoma and nontumor (BRL3A) cells treated or not treated with the compound. Note the normal arrangement of tubulin and actin fibers in the control cells; the formation of monopolar spindles in MCF-7, HS58T and BRL3A cells; and the formation of tripolar spindles in T47D cells. Figure from Militão et al. (9797. Tesauro C, Fiorani P, D’Annessa I, Chillemi G, Turchi G, Desideri A, Erybraedin C. A natural compound from the plant Bituminaria bituminosa, inhibits both the cleavage and religation activities of human topoisomerase I. Biochem J. 2010;425(3):531-9, http://dx.doi.org/10.1042/BJ20091127.
http://dx.doi.org/10.1042/BJ20091127...
).

CONCLUSION

Antimitotic chemotherapeutics such as Taxol (paclitaxel), a microtubule-stabilizing agent that causes mitotic arrest, are currently successfully employed in cancer treatment (107107. Weaver BA. How Taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25(18):2677-81, http://dx.doi.org/10.1091/mbc.e14-04-0916.
http://dx.doi.org/10.1091/mbc.e14-04-091...
). Regardless of the relative success of antimitotics, however, the death rate from cancer remains unacceptably high worldwide, and many efforts on different fronts have been made to improve cancer therapy (108108. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359-86, http://dx.doi.org/10.1002/ijc.29210.
http://dx.doi.org/10.1002/ijc.29210...
). The reduction of drug resistance and side effects associated with antimitotics, such as neurotoxicity and bone marrow suppression, is a necessary improvement (6060. Doménech E, Malumbres M. Mitosis-targeting therapies: a troubleshooting guide. Curr Opin Pharmacol. 2013;13(4):519-28, http://dx.doi.org/10.1016/j.coph.2013.03.011.
http://dx.doi.org/10.1016/j.coph.2013.03...
61. Schöffski P, Awada A, Dumez H, Gil T, Bartholomeus S, Wolter P, et al. A phase I, dose-escalation study of the novel Polo-like kinase inhibitor volasertib (BI 6727) in patients with advanced solid tumours. Eur J Cancer. 2012;48(2):179-86, http://dx.doi.org/10.1016/j.ejca.2011.11.001.
http://dx.doi.org/10.1016/j.ejca.2011.11...
62. Arkenau HT, Plummer R, Molife LR, Olmos D, Yap TA, Squires M, et al. A phase I dose escalation study of AT9283, a small molecule inhibitor of aurora kinases, in patients with advanced solid malignancies. Ann Oncol. 2012;23(5):1307-13, http://dx.doi.org/10.1093/annonc/mdr451.
http://dx.doi.org/10.1093/annonc/mdr451...
-6363. Jones R, Vuky J, Elliott T, Mead G, Arranz JA, Chester J, et al. Phase II study to assess the efficacy, safety and tolerability of the mitotic spindle kinesin inhibitor AZD4877 in patients with recurrent advanced urothelial cancer. Invest New Drugs. 2013;31(4):1001-7, http://dx.doi.org/10.1007/s10637-013-9926-y.
http://dx.doi.org/10.1007/s10637-013-992...
). In this review, we presented new antimitotic classes investigated by our research group that address the main issues of current chemotherapeutics.

The combretastatin derivatives are analogs of CA-4, an inhibitor of tubulin polymerization and microtubule formation (6868. Combes S, Barbier P, Douillard S, McLeer-Florin A, Bourgarel-Rey V, Pierson JT, et al. Synthesis and biological evaluation of 4-arylcoumarin analogues of combretastatins. Part 2. J Med Chem. 2011;54(9):3153-62, http://dx.doi.org/10.1021/jm901826e.
http://dx.doi.org/10.1021/jm901826e...
). Because the functions of tubulin and microtubules are not limited to mitosis, CA-4 may affect quiescent or interphase cells, likely causing side effects in normal tissues (109109. Shi YW, Yuan W, Wang X, Gong J, Zhu SX, Chai LL, et al. Combretastatin A-4 efficiently inhibits angiogenesis and induces neuronal apoptosis in zebrafish. Sci Rep. 2016;6(1):30189, http://dx.doi.org/10.1038/srep30189.
http://dx.doi.org/10.1038/srep30189...
). Thus, our structure–activity relationship studies identified structural characteristics of improved CA-4 analogs. We showed that the stability issues related to the cis stilbene nature of the combretastatin core were overcome by the replacement of the cis olefin double bond with an NAH framework, whose variation may result in inactive compounds. We discovered that compounds with the aromatic rings (A and B) in a trans configuration maintain tubulin-binding activity. Last, we showed a direct relationship between the increase in hydrophobicity and the cytotoxic effect of CA-4 analogs. Therefore, we described combretastatin analogs with increased cytotoxicity and selectivity toward tumor cells that can be lead molecules for new antimitotic agents inducing fewer side effects.

We also reviewed antimitotics derived from the natural products withanolides and pterocarpans. Indeed, natural sources are major sources of lead molecules and new drugs (110110. Newman DJ, Cragg GM. Natural Products as Sources of New Drugs from 1981 to 2014. J Nat Prod. 2016;79(3):629-61, http://dx.doi.org/10.1021/acs.jnatprod.5b01055.
http://dx.doi.org/10.1021/acs.jnatprod.5...
) such as CA-4 and Taxol. Although the mechanism of action of withanolides (withaphysalin F and withaferin A) is not fully understood, the targets of these agents may be proteins functioning in interphase, such as β-tubulin, vimentin, annexin II, heat shock protein 90 and proteasome subunits (8181. Antony ML, Lee J, Hahm ER, Kim SH, Marcus AI, Kumari V, et al. Growth arrest by the antitumor steroidal lactone withaferin A in human breast cancer cells is associated with down-regulation and covalent binding at cysteine 303 of β-tubulin. J Biol Chem. 2014;289(3):1852-65, http://dx.doi.org/10.1074/jbc.M113.496844.
http://dx.doi.org/10.1074/jbc.M113.49684...
,8585. Bargagna-Mohan P, Hamza A, Kim YE, Khuan Abby Ho Y, Mor-Vaknin N, Wendschlag N, et al. The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. Chem Biol. 2007;14(6):623-34, http://dx.doi.org/10.1016/j.chembiol.2007.04.010.
http://dx.doi.org/10.1016/j.chembiol.200...
86. Falsey RR, Marron MT, Gunaherath GM, Shirahatti N, Mahadevan D, Gunatilaka AA, et al. Actin microfilament aggregation induced by withaferin A is mediated by annexin II. Nat Chem Biol. 2006;2(1):33-8, http://dx.doi.org/10.1038/nchembio755.
http://dx.doi.org/10.1038/nchembio755...
87. Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B, et al. Withaferin A targets heat shock protein 90 in pancreatic cancer cells. Biochem Pharmacol. 2010;79(4):542-51, http://dx.doi.org/10.1016/j.bcp.2009.09.017.
http://dx.doi.org/10.1016/j.bcp.2009.09....
-8888. Yang H, Shi G, Dou QP. The tumor proteasome is a primary target for the natural anticancer compound Withaferin A isolated from “Indian winter cherry”. Mol Pharmacol. 2007;71(2):426-37, http://dx.doi.org/10.1124/mol.106.030015.
http://dx.doi.org/10.1124/mol.106.030015...
). Although these proteins are not mitosis-specific, their modulation may disrupt the cell cycle. Thus, withanolides may have greater-than-expected activity against targets of cancer cells in interphase (5353. Komlodi-Pasztor E, Sackett DL, Fojo AT. Inhibitors targeting mitosis: tales of how great drugs against a promising target were brought down by a flawed rationale. Clin Cancer Res. 2012;18(1):51-63, http://dx.doi.org/10.1158/1078-0432.CCR-11-0999.
http://dx.doi.org/10.1158/1078-0432.CCR-...
,5454. Mitchison TJ. The proliferation rate paradox in antimitotic chemotherapy. Mol Biol Cell. 2012;23(1):1-6, http://dx.doi.org/10.1091/mbc.e10-04-0335.
http://dx.doi.org/10.1091/mbc.e10-04-033...
), which could decrease the development of resistance to chemotherapy.

Finally, (+)2,3,9-trimethoxypterocarpan is an antimitotic agent that induces cell cycle arrest at the G2/M phase transition in leukemic cells and causes the aberrant organization of mitotic spindle microtubules in breast cancer cells. Although treatment of cells in M phase was found to result in monopolar and tripolar mitotic spindles, cells treated in interphase exhibited no microtubule modifications (102102. Militão GC, Prado MP, Pessoa C, de Moraes MO, Silveira ER, Lima MA, et al. Pterocarpans induce tumor cell death through persistent mitotic arrest during prometaphase. Biochimie. 2014;104:147-55, http://dx.doi.org/10.1016/j.biochi.2014.06.005.
http://dx.doi.org/10.1016/j.biochi.2014....
). These characteristics resemble the phenotype of cells treated with monastrol, a specific inhibitor of the kinesin motor protein Eg5, which is expressed only during mitosis (1010. Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL, Mitchison TJ. Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science. 1999;286(5441):971-4, http://dx.doi.org/10.1126/science.286.5441.971.
http://dx.doi.org/10.1126/science.286.54...
,1111. Jackson JR, Patrick DR, Dar MM, Huang PS. Targeted anti-mitotic therapies: can we improve on tubulin agents? Nat Rev Cancer. 2007;7(2):107-17, http://dx.doi.org/10.1038/nrc2049.
http://dx.doi.org/10.1038/nrc2049...
). Such lines of evidence suggest that the target of (+)2,3,9-trimethoxypterocarpan is a mitosis-specific protein, and therefore, the action of this compound may be restricted to cells in mitosis. Therefore, this compound could potentially spare normal quiescent and interphase cells during clinical treatment, preventing neurotoxicity, for example.

All compounds reviewed demonstrate the potential to address important issues regarding current antimitotic-based cancer therapy. Therefore, in vivo antitumor assays will be performed to confirm these antimitotics as valuable lead molecules for the development of new anticancer drugs. However, specificity and toxicity issues concerning such molecules may arise but can be solved by combining antimitotics with other antineoplastic agents and/or monoclonal antibodies designed to bind to surface tumor antigens. This strategy can be implemented through direct covalent conjugation or drug encapsulation in nanoparticles functionalized with antibodies. Antibody-drug conjugates are a new trend in anticancer research (111111. Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, et al. Antibody-Drug Conjugates for Cancer Therapy: Chemistry to Clinical Implications. Pharmaceuticals (Basel). 2018;11(2). pii: E32, http://dx.doi.org/10.3390/ph11020032.
http://dx.doi.org/10.3390/ph11020032...
); this approach also spurs the search for new and improved antimitotics.

ACKNOWLEDGMENTS

We are grateful to the Brazilian Coordination of Improvement of Higher Education Personnel (CAPES) and the Brazilian National Council for Scientific and Technological Development (CNPq) for the financial support for works cited in sections 4, 5 and 6. Dr. A. Leyva (USA) provided English language editing of the manuscript.

REFERENCES

  • 1
    Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74, http://dx.doi.org/10.1016/j.cell.2011.02.013
    » http://dx.doi.org/10.1016/j.cell.2011.02.013
  • 2
    Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9(3):153-66, http://dx.doi.org/10.1038/nrc2602
    » http://dx.doi.org/10.1038/nrc2602
  • 3
    Marzo I, Naval J. Antimitotic drugs in cancer chemotherapy: promises and pitfalls. Biochem Pharmacol. 2013;86(6):703-10, http://dx.doi.org/10.1016/j.bcp.2013.07.010
    » http://dx.doi.org/10.1016/j.bcp.2013.07.010
  • 4
    Sudakin V, Yen TJ. Targeting mitosis for anti-cancer therapy. BioDrugs. 2007;21(4):225-33, http://dx.doi.org/10.2165/00063030-200721040-00003
    » http://dx.doi.org/10.2165/00063030-200721040-00003
  • 5
    Penna LS, Henriques JAP, Bonatto D. Anti-mitotic agents: are they emerging molecules for cancer treatment? Pharmacol Ther. 2017;173:67-82, http://dx.doi.org/10.1016/j.pharmthera.2017.02.007
    » http://dx.doi.org/10.1016/j.pharmthera.2017.02.007
  • 6
    Muroyama A, Lechler T. Microtubule organization, dynamics and functions in differentiated cells. Development. 2017;144(17):3012-21, http://dx.doi.org/10.1242/dev.153171
    » http://dx.doi.org/10.1242/dev.153171
  • 7
    Harrison MR, Holen KD, Liu G. Beyond taxanes: a review of novel agents that target mitotic tubulin and microtubules, kinases, and kinesins. Clin Adv Hematol Oncol. 2009;7(1):54-64.
  • 8
    Campos SM, Dizon DS. Antimitotic inhibitors. Hematol Oncol Clin North Am. 2012;26(3):607-28, http://dx.doi.org/10.1016/j.hoc.2012.01.007
    » http://dx.doi.org/10.1016/j.hoc.2012.01.007
  • 9
    Gascoigne KE, Taylor SS. How do anti-mitotic drugs kill cancer cells? J Cell Sci. 2009;122(Pt 15):2579-85, http://dx.doi.org/10.1242/jcs.039719
    » http://dx.doi.org/10.1242/jcs.039719
  • 10
    Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL, Mitchison TJ. Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science. 1999;286(5441):971-4, http://dx.doi.org/10.1126/science.286.5441.971
    » http://dx.doi.org/10.1126/science.286.5441.971
  • 11
    Jackson JR, Patrick DR, Dar MM, Huang PS. Targeted anti-mitotic therapies: can we improve on tubulin agents? Nat Rev Cancer. 2007;7(2):107-17, http://dx.doi.org/10.1038/nrc2049
    » http://dx.doi.org/10.1038/nrc2049
  • 12
    Myers SM, Collins I. Recent findings and future directions for interpolar mitotic kinesin inhibitors in cancer therapy. Future Med Chem. 2016;8(4):463-89, http://dx.doi.org/10.4155/fmc.16.5
    » http://dx.doi.org/10.4155/fmc.16.5
  • 13
    Kung PP, Martinez R, Zhu Z, Zager M, Blasina A, Rymer I, et al. Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer. Mol Cancer Ther. 2014;13(8):2104-15, http://dx.doi.org/10.1158/1535-7163.MCT-14-0083-T
    » http://dx.doi.org/10.1158/1535-7163.MCT-14-0083-T
  • 14
    Henderson MC, Shaw YJ, Wang H, Han H, Hurley LH, Flynn G, et al. UA62784, a novel inhibitor of centromere protein E kinesin-like protein. Mol Cancer Ther. 2009;8(1):36-44, http://dx.doi.org/10.1158/1535-7163.MCT-08-0789
    » http://dx.doi.org/10.1158/1535-7163.MCT-08-0789
  • 15
    Ohashi A, Ohori M, Iwai K, Nambu T, Miyamoto M, Kawamoto T, et al. A Novel Time-Dependent CENP-E Inhibitor with Potent Antitumor Activity. PLoS One. 2015;10(12):e0144675, http://dx.doi.org/10.1371/journal.pone.0144675
    » http://dx.doi.org/10.1371/journal.pone.0144675
  • 16
    Chung V, Heath EI, Schelman WR, Johnson BM, Kirby LC, Lynch KM, et al. First-time-in-human study of GSK923295, a novel antimitotic inhibitor of centromere-associated protein E (CENP-E), in patients with refractory cancer. Cancer Chemother Pharmacol. 2012;69(3):733-41, http://dx.doi.org/10.1007/s00280-011-1756-z
    » http://dx.doi.org/10.1007/s00280-011-1756-z
  • 17
    Hegde PS, Cogswell J, Carrick K, Jackson J, Wood KW, Eng WK, et al. Differential gene expression analysis of kinesin spindle protein in human solid tumors. Proc Am Soc Clin Oncol. 2003;22:535.
  • 18
    Maliga Z, Junqueira M, Toyoda Y, Ettinger A, Mora-Bermúdez F, Klemm RW, et al. A genomic toolkit to investigate kinesin and myosin motor function in cells. Nat Cell Biol. 2013;15(3):325-34, http://dx.doi.org/10.1038/ncb2689
    » http://dx.doi.org/10.1038/ncb2689
  • 19
    Pitts TM, Davis SL, Eckhardt SG, Bradshaw-Pierce EL. Targeting nuclear kinases in cancer: development of cell cycle kinase inhibitors. Pharmacol Ther. 2014;142(2):258-69, http://dx.doi.org/10.1016/j.pharmthera.2013.12.010
    » http://dx.doi.org/10.1016/j.pharmthera.2013.12.010
  • 20
    Bartek J, Lukas J. Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell. 2003;3(5):421-9, http://dx.doi.org/10.1016/S1535-6108(03)00110-7
    » http://dx.doi.org/10.1016/S1535-6108(03)00110-7
  • 21
    Ortega S, Malumbres M, Barbacid M. Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta. 2002;1602(1):73-87, http://dx.doi.org/10.1016/S0304-419X(02)00037-9
    » http://dx.doi.org/10.1016/S0304-419X(02)00037-9
  • 22
    Cirillo D, Pentimalli F, Giordano A. Peptides or small molecules? Different approaches to develop more effective CDK inhibitors. Curr Med Chem. 2011;18(19):2854-66, http://dx.doi.org/10.2174/092986711796150496
    » http://dx.doi.org/10.2174/092986711796150496
  • 23
    Canavese M, Santo L, Raje N. Cyclin dependent kinases in cancer: potential for therapeutic intervention. Cancer Biol Ther. 2012;13(7):451-7, http://dx.doi.org/10.4161/cbt.19589
    » http://dx.doi.org/10.4161/cbt.19589
  • 24
    Roshak AK, Capper EA, Imburgia C, Fornwald J, Scott G, Marshall LA. The human polo-like kinase, PLK, regulates cdc2/cyclin B through phosphorylation and activation of the cdc25C phosphatase. Cell Signal. 2000;12(6):405-11, http://dx.doi.org/10.1016/S0898-6568(00)00080-2
    » http://dx.doi.org/10.1016/S0898-6568(00)00080-2
  • 25
    Tut TG, Lim SH, Dissanayake IU, Descallar J, Chua W, Ng W, et al. Upregulated Polo-like Kinase 1 expression correlates with inferior survival outcomes in rectal cancer. PLoS One. 2015;10(6):e0129313, http://dx.doi.org/10.1371/journal.pone.0129313
    » http://dx.doi.org/10.1371/journal.pone.0129313
  • 26
    Kanaji S, Saito H, Tsujitani S, Matsumoto S, Tatebe S, Kondo A, et al. Expression of polo-like kinase 1 (PLK1) protein predicts the survival of patients with gastric carcinoma. Oncology. 2006;70(2):126-33, http://dx.doi.org/10.1159/000093003
    » http://dx.doi.org/10.1159/000093003
  • 27
    Gumireddy K, Reddy MV, Cosenza SC, Boominathan R, Baker SJ, Papathi N, et al. ON01910, a non-ATP-competitive small molecule inhibitor of Plk1, is a potent anticancer agent. Cancer Cell. 2005;7(3):275-86, http://dx.doi.org/10.1016/j.ccr.2005.02.009
    » http://dx.doi.org/10.1016/j.ccr.2005.02.009
  • 28
    Rudolph D, Steegmaier M, Hoffmann M, Grauert M, Baum A, Quant J, et al. BI 6727, a Polo-like kinase inhibitor with improved pharmacokinetic profile and broad antitumor activity. Clin Cancer Res. 2009;15(9):3094-102, http://dx.doi.org/10.1158/1078-0432.CCR-08-2445
    » http://dx.doi.org/10.1158/1078-0432.CCR-08-2445
  • 29
    Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel therapy targets in cancers. Oncotarget. 2017;8(14):23937-54, http://dx.doi.org/10.18632/oncotarget.14893
    » http://dx.doi.org/10.18632/oncotarget.14893
  • 30
    Cirak Y, Furuncuoglu Y, Yapicier O, Aksu A, Cubukcu E. Aurora A overexpression in breast cancer patients induces taxane resistance and results in worse prognosis. J BUON. 2015;20(6):1414-9.
  • 31
    Davidson B, Nymoen DA, Elgaaen BV, Staff AC, Tropé CG, Kærn J, et al. BUB1 mRNA is significantly co-expressed with AURKA and AURKB mRNA in advanced-stage ovarian serous carcinoma. Virchows Arch. 2014;464(6):701-7, http://dx.doi.org/10.1007/s00428-014-1577-7
    » http://dx.doi.org/10.1007/s00428-014-1577-7
  • 32
    Jeng YM, Peng SY, Lin CY, Hsu HC. Overexpression and amplification of Aurora-A in hepatocellular carcinoma. Clin Cancer Res. 2004;10(6):2065-71, http://dx.doi.org/10.1158/1078-0432.CCR-1057-03
    » http://dx.doi.org/10.1158/1078-0432.CCR-1057-03
  • 33
    Heredia FF, de Sousa JC, Ribeiro Junior HL, Carvalho AF, Magalhães SM, Pinheiro RF. Proteins related to the spindle and checkpoint mitotic emphasize the different pathogenesis of hypoplastic MDS. Leuk Res. 2014;38(2):218-24, http://dx.doi.org/10.1016/j.leukres.2013.11.003
    » http://dx.doi.org/10.1016/j.leukres.2013.11.003
  • 34
    Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov. 2009;8(7):547-66, http://dx.doi.org/10.1038/nrd2907
    » http://dx.doi.org/10.1038/nrd2907
  • 35
    Matulonis UA, Sharma S, Ghamande S, Gordon MS, Del Prete SA, Ray-Coquard I, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol. 2012;127(1):63-9, http://dx.doi.org/10.1016/j.ygyno.2012.06.040
    » http://dx.doi.org/10.1016/j.ygyno.2012.06.040
  • 36
    Yang J, Ikezoe T, Nishioka C, Tasaka T, Taniguchi A, Kuwayama Y, et al. AZD1152, a novel and selective aurora B kinase inhibitor, induces growth arrest, apoptosis, and sensitization for tubulin depolymerizing agent or topoisomerase II inhibitor in human acute leukemia cells in vitro and in vivo Blood. 2007;110(6):2034-40, http://dx.doi.org/10.1182/blood-2007-02-073700
    » http://dx.doi.org/10.1182/blood-2007-02-073700
  • 37
    Giles FJ, Cortes J, Jones D, Bergstrom D, Kantarjian H, Freedman SJ. MK-0457, a novel kinase inhibitor, is active in patients with chronic myeloid leukemia or acute lymphocytic leukemia with the T315I BCR-ABL mutation. Blood. 2007;109(2):500-2, http://dx.doi.org/10.1182/blood-2006-05-025049
    » http://dx.doi.org/10.1182/blood-2006-05-025049
  • 38
    Guo K, Shelat AA, Guy RK, Kastan MB. Development of a cell-based, high-throughput screening assay for ATM kinase inhibitors. J Biomol Screen. 2014;19(4):538-46, http://dx.doi.org/10.1177/1087057113520325
    » http://dx.doi.org/10.1177/1087057113520325
  • 39
    Lin K, Adamson J, Johnson GG, Carter A, Oates M, Wade R, et al. Functional analysis of the ATM-p53-p21 pathway in the LRF CLL4 trial: blockade at the level of p21 is associated with short response duration. Clin Cancer Res. 2012;18(15):4191-200, http://dx.doi.org/10.1158/1078-0432.CCR-11-2936
    » http://dx.doi.org/10.1158/1078-0432.CCR-11-2936
  • 40
    Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, et al. COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43(Database issue):D805-11, http://dx.doi.org/10.1093/nar/gku1075
    » http://dx.doi.org/10.1093/nar/gku1075
  • 41
    Choi M, Kipps T, Kurzrock R. ATM Mutations in Cancer: therapeutic Implications. Mol Cancer Ther. 2016;15(8):1781-91, http://dx.doi.org/10.1158/1535-7163.MCT-15-0945
    » http://dx.doi.org/10.1158/1535-7163.MCT-15-0945
  • 42
    Magnussen GI, Hellesylt E, Nesland JM, Trope CG, Flørenes VA, Holm R. High expression of wee1 is associated with malignancy in vulvar squamous cell carcinoma patients. BMC Cancer. 2013;13(1):288, http://dx.doi.org/10.1186/1471-2407-13-288
    » http://dx.doi.org/10.1186/1471-2407-13-288
  • 43
    Porter CC, Kim J, Fosmire S, Gearheart CM, van Linden A, Baturin D, et al. Integrated genomic analyses identify WEE1 as a critical mediator of cell fate and a novel therapeutic target in acute myeloid leukemia. Leukemia. 2012;26(6):1266-76, http://dx.doi.org/10.1038/leu.2011.392
    » http://dx.doi.org/10.1038/leu.2011.392
  • 44
    Slipicevic A, Holth A, Hellesylt E, Tropé CG, Davidson B, Flørenes VA. Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. Gynecol Oncol. 2014;135(1):118-24, http://dx.doi.org/10.1016/j.ygyno.2014.07.102
    » http://dx.doi.org/10.1016/j.ygyno.2014.07.102
  • 45
    Wang Y, Li J, Booher RN, Kraker A, Lawrence T, Leopold WR, et al. Radiosensitization of p53 mutant cells by PD0166285, a novel G(2) checkpoint abrogator. Cancer Res. 2001;61(22):8211-7.
  • 46
    Palmer BD, Thompson AM, Booth RJ, Dobrusin EM, Kraker AJ, Lee HH, et al. 4-Phenylpyrrolo[3,4-c]carbazole-1,3(2H,6H)-dione inhibitors of the checkpoint kinase Wee1. Structure-activity relationships for chromophore modification and phenyl ring substitution. J Med Chem. 2006;49(16):4896-911, http://dx.doi.org/10.1021/jm0512591
    » http://dx.doi.org/10.1021/jm0512591
  • 47
    Matheson CJ, Backos DS, Reigan P. Targeting WEE1 Kinase in Cancer. Trends Pharmacol Sci. 2016;37(10):872-81, http://dx.doi.org/10.1016/j.tips.2016.06.006
    » http://dx.doi.org/10.1016/j.tips.2016.06.006
  • 48
    Grigoreva TA, Tribulovich VG, Garabadzhiu AV, Melino G, Barlev NA. The 26S proteasome is a multifaceted target for anti-cancer therapies. Oncotarget. 2015;6(28):24733-49, http://dx.doi.org/10.18632/oncotarget.4619
    » http://dx.doi.org/10.18632/oncotarget.4619
  • 49
    Moreau P. The emerging role of carfilzomib combination therapy in the management of multiple myeloma. Expert Rev Hematol. 2014;7(2):265-90, http://dx.doi.org/10.1586/17474086.2014.873699
    » http://dx.doi.org/10.1586/17474086.2014.873699
  • 50
    Teicher BA, Tomaszewski JE, Tomaszewski JE. Proteasome inhibitors. Biochem Pharmacol. 2015;96(1):1-9, http://dx.doi.org/10.1016/j.bcp.2015.04.008
    » http://dx.doi.org/10.1016/j.bcp.2015.04.008
  • 51
    Selvam C, Jordan BC, Prakash S, Mutisya D, Thilagavathi R. Pterocarpan scaffold: A natural lead molecule with diverse pharmacological properties. Eur J Med Chem. 2017;128:219-36, http://dx.doi.org/10.1016/j.ejmech.2017.01.023
    » http://dx.doi.org/10.1016/j.ejmech.2017.01.023
  • 52
    Chan KS, Koh CG, Li HY. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis. 2012;3(10):e411, http://dx.doi.org/10.1038/cddis.2012.148
    » http://dx.doi.org/10.1038/cddis.2012.148
  • 53
    Komlodi-Pasztor E, Sackett DL, Fojo AT. Inhibitors targeting mitosis: tales of how great drugs against a promising target were brought down by a flawed rationale. Clin Cancer Res. 2012;18(1):51-63, http://dx.doi.org/10.1158/1078-0432.CCR-11-0999
    » http://dx.doi.org/10.1158/1078-0432.CCR-11-0999
  • 54
    Mitchison TJ. The proliferation rate paradox in antimitotic chemotherapy. Mol Biol Cell. 2012;23(1):1-6, http://dx.doi.org/10.1091/mbc.e10-04-0335
    » http://dx.doi.org/10.1091/mbc.e10-04-0335
  • 55
    Manchado E, Guillamot M, Malumbres M. Killing cells by targeting mitosis. Cell Death Differ. 2012;19(3):369-77, http://dx.doi.org/10.1038/cdd.2011.197
    » http://dx.doi.org/10.1038/cdd.2011.197
  • 56
    Freedman H, Huzil JT, Luchko T, Ludueãa RF, Tuszynski JA. Identification and characterization of an intermediate taxol binding site within microtubule nanopores and a mechanism for tubulin isotype binding selectivity. J Chem Inf Model. 2009;49(2):424-36, http://dx.doi.org/10.1021/ci8003336
    » http://dx.doi.org/10.1021/ci8003336
  • 57
    Ferrandina G, Zannoni GF, Martinelli E, Paglia A, Gallotta V, Mozzetti S, et al. Class III β-tubulin overexpression is a marker of poor clinical outcome in advanced ovarian cancer patients. Clin Cancer Res. 2006;12(9):2774-9, http://dx.doi.org/10.1158/1078-0432.CCR-05-2715
    » http://dx.doi.org/10.1158/1078-0432.CCR-05-2715
  • 58
    Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist. 2003;8(5):411-24, http://dx.doi.org/10.1634/theoncologist.8-5-411
    » http://dx.doi.org/10.1634/theoncologist.8-5-411
  • 59
    Fojo AT, Menefee M. Microtubule targeting agents: basic mechanisms of multidrug resistance (MDR). Semin Oncol. 2005;32(6 Suppl 7):S3-8, http://dx.doi.org/10.1053/j.seminoncol.2005.09.010
    » http://dx.doi.org/10.1053/j.seminoncol.2005.09.010
  • 60
    Doménech E, Malumbres M. Mitosis-targeting therapies: a troubleshooting guide. Curr Opin Pharmacol. 2013;13(4):519-28, http://dx.doi.org/10.1016/j.coph.2013.03.011
    » http://dx.doi.org/10.1016/j.coph.2013.03.011
  • 61
    Schöffski P, Awada A, Dumez H, Gil T, Bartholomeus S, Wolter P, et al. A phase I, dose-escalation study of the novel Polo-like kinase inhibitor volasertib (BI 6727) in patients with advanced solid tumours. Eur J Cancer. 2012;48(2):179-86, http://dx.doi.org/10.1016/j.ejca.2011.11.001
    » http://dx.doi.org/10.1016/j.ejca.2011.11.001
  • 62
    Arkenau HT, Plummer R, Molife LR, Olmos D, Yap TA, Squires M, et al. A phase I dose escalation study of AT9283, a small molecule inhibitor of aurora kinases, in patients with advanced solid malignancies. Ann Oncol. 2012;23(5):1307-13, http://dx.doi.org/10.1093/annonc/mdr451
    » http://dx.doi.org/10.1093/annonc/mdr451
  • 63
    Jones R, Vuky J, Elliott T, Mead G, Arranz JA, Chester J, et al. Phase II study to assess the efficacy, safety and tolerability of the mitotic spindle kinesin inhibitor AZD4877 in patients with recurrent advanced urothelial cancer. Invest New Drugs. 2013;31(4):1001-7, http://dx.doi.org/10.1007/s10637-013-9926-y
    » http://dx.doi.org/10.1007/s10637-013-9926-y
  • 64
    Fürst R, Zupkó I, Berényi A, Ecker GF, Rinner U. Synthesis and antitumor-evaluation of cyclopropyl-containing combretastatin analogs. Bioorg Med Chem Lett. 2009;19(24):6948-51, http://dx.doi.org/10.1016/j.bmcl.2009.10.064
    » http://dx.doi.org/10.1016/j.bmcl.2009.10.064
  • 65
    Shan Y, Zhang J, Liu Z, Wang M, Dong Y. Developments of combretastatin A-4 derivatives as anticancer agents. Curr Med Chem. 2011;18(4):523-38, http://dx.doi.org/10.2174/092986711794480221
    » http://dx.doi.org/10.2174/092986711794480221
  • 66
    Seddigi ZS, Malik MS, Saraswati AP, Ahmed SA, Babalghith AO, Lamfon HA, et al. Recent advances in combretastatin based derivatives and prodrugs as antimitotic agents. MedChemComm. 2017;8(8):1592-603, http://dx.doi.org/10.1039/C7MD00227K
    » http://dx.doi.org/10.1039/C7MD00227K
  • 67
    Yang T, Wang Y, Li Z, Dai W, Yin J, Liang L, et al. Targeted delivery of a combination therapy consisting of combretastatin A4 and low-dose doxorubicin against tumor neovasculature. Nanomedicine (Lond). 2012;8(1):81-92, http://dx.doi.org/10.1016/j.nano.2011.05.003
    » http://dx.doi.org/10.1016/j.nano.2011.05.003
  • 68
    Combes S, Barbier P, Douillard S, McLeer-Florin A, Bourgarel-Rey V, Pierson JT, et al. Synthesis and biological evaluation of 4-arylcoumarin analogues of combretastatins. Part 2. J Med Chem. 2011;54(9):3153-62, http://dx.doi.org/10.1021/jm901826e
    » http://dx.doi.org/10.1021/jm901826e
  • 69
    Duarte CD, Barreiro EJ, Fraga CA. Privileged structures: a useful concept for the rational design of new lead drug candidates. Mini Rev Med Chem. 2007;7(11):1108-19, http://dx.doi.org/10.2174/138955707782331722
    » http://dx.doi.org/10.2174/138955707782331722
  • 70
    do Amaral DN, Cavalcanti BC, Bezerra DP, Ferreira PM, Castro RP, Sabino JR, et al. Docking, synthesis and antiproliferative activity of N-acylhydrazone derivatives designed as combretastatin A4 analogues. PLoS One. 2014;9(3):e85380, http://dx.doi.org/10.1371/journal.pone.0085380
    » http://dx.doi.org/10.1371/journal.pone.0085380
  • 71
    Hu L, Li ZR, Li Y, Qu J, Ling YH, Jiang JD, et al. Synthesis and structure-activity relationships of carbazole sulfonamides as a novel class of antimitotic agents against solid tumors. J Med Chem. 2006;49(21):6273-82, http://dx.doi.org/10.1021/jm060546h
    » http://dx.doi.org/10.1021/jm060546h
  • 72
    Cavalcanti BC, Barros FW, Cabral IO, Ferreira JR, Magalhães HI, Júnior HV, et al. Preclinical genotoxicology of nor-β-lapachone in human cultured lymphocytes and Chinese hamster lung fibroblasts. Chem Res Toxicol. 2011;24(9):1560-74, http://dx.doi.org/10.1021/tx200180y
    » http://dx.doi.org/10.1021/tx200180y
  • 73
    Carneiro TR, Amaral DN, Fokoue HH, Sant’Anna CM, Porras ML, Oliveira AC. Synthesis, pharmacological evaluation and docking study of a new modulator of microtubule polymerization. Lett Drug Design Disc. 2018;15(7):778-86, http://dx.doi.org/10.2174/1570180814666171012162557
    » http://dx.doi.org/10.2174/1570180814666171012162557
  • 74
    Glotter E. Withanolides and related ergostane-type steroids. Nat Prod Rep. 1991;8(4):415-40, http://dx.doi.org/10.1039/np9910800415
    » http://dx.doi.org/10.1039/np9910800415
  • 75
    Zhu F, Dai C, Fu Y, Loo JF, Xia D, Gao SP, et al. Physalin A exerts anti-tumor activity in non-small cell lung cancer cell lines by suppressing JAK/STAT3 signaling. Oncotarget. 2016;7(8):9462-76, http://dx.doi.org/10.18632/oncotarget.7051
    » http://dx.doi.org/10.18632/oncotarget.7051
  • 76
    Rocha DD, Militão GC, Veras ML, Pessoa OD, Silveira ER, Alves AP, et al. Selective cytotoxicity of withaphysalins in myeloid leukemia cell lines versus peripheral blood mononuclear cells. Life Sci. 2006;79(18):1692-701, http://dx.doi.org/10.1016/j.lfs.2006.05.026
    » http://dx.doi.org/10.1016/j.lfs.2006.05.026
  • 77
    Kour K, Pandey A, Suri KA, Satti NK, Gupta KK, Bani S. Restoration of stress-induced altered T cell function and corresponding cytokines patterns by Withanolide A. Int Immunopharmacol. 2009;9(10):1137-44, http://dx.doi.org/10.1016/j.intimp.2009.05.011
    » http://dx.doi.org/10.1016/j.intimp.2009.05.011
  • 78
    Mareggiani G, Picollo MI, Zerba E, Burton G, Tettamanzi MC, Benedetti-Doctorovich MO, et al. Antifeedant activity of withanolides from Salpichroa origanifolia on Musca domestica. J Nat Prod. 2000;63(8):1113-6, http://dx.doi.org/10.1021/np0001068
    » http://dx.doi.org/10.1021/np0001068
  • 79
    Soares MB, Brustolim D, Santos LA, Bellintani MC, Paiva FP, Ribeiro YM, et al. Physalins B, F and G, seco-steroids purified from Physalis angulata L., inhibit lymphocyte function and allogeneic transplant rejection. Int Immunopharmacol. 2006;6(3):408-14, http://dx.doi.org/10.1016/j.intimp.2005.09.007
    » http://dx.doi.org/10.1016/j.intimp.2005.09.007
  • 80
    Shanazbanu S, Shashidara S, Babu VL, Dhanapal R. Isolation of withaferin A from Withania somnifera Dun leaves and its antibacterial activity. Asian J Chem. 2006;18(2):1243-7.
  • 81
    Antony ML, Lee J, Hahm ER, Kim SH, Marcus AI, Kumari V, et al. Growth arrest by the antitumor steroidal lactone withaferin A in human breast cancer cells is associated with down-regulation and covalent binding at cysteine 303 of β-tubulin. J Biol Chem. 2014;289(3):1852-65, http://dx.doi.org/10.1074/jbc.M113.496844
    » http://dx.doi.org/10.1074/jbc.M113.496844
  • 82
    Das T, Roy KS, Chakrabarti T, Mukhopadhyay S, Roychoudhury S. Withaferin A modulates the Spindle assembly checkpoint by degradation of Mad2-Cdc20 complex in colorectal cancer cell lines. Biochem Pharmacol. 2014;91(1):31-9, http://dx.doi.org/10.1016/j.bcp.2014.06.022
    » http://dx.doi.org/10.1016/j.bcp.2014.06.022
  • 83
    Roy RV, Suman S, Das TP, Luevano JE, Damodaran C. Withaferin-A induces mitotic catastrophe and growth arrest in prostate cancer cells. J Nat Prod. 2013;76(10):1909-15, http://dx.doi.org/10.1021/np400441f
    » http://dx.doi.org/10.1021/np400441f
  • 84
    Stan SD, Zeng Y, Singh SV. Ayurvedic medicine constituent withaferin a causes G2 and M phase cell cycle arrest in human breast cancer cells. Nutr Cancer. 2008;60(Suppl 1):51-60, http://dx.doi.org/10.1080/01635580802381477
    » http://dx.doi.org/10.1080/01635580802381477
  • 85
    Bargagna-Mohan P, Hamza A, Kim YE, Khuan Abby Ho Y, Mor-Vaknin N, Wendschlag N, et al. The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. Chem Biol. 2007;14(6):623-34, http://dx.doi.org/10.1016/j.chembiol.2007.04.010
    » http://dx.doi.org/10.1016/j.chembiol.2007.04.010
  • 86
    Falsey RR, Marron MT, Gunaherath GM, Shirahatti N, Mahadevan D, Gunatilaka AA, et al. Actin microfilament aggregation induced by withaferin A is mediated by annexin II. Nat Chem Biol. 2006;2(1):33-8, http://dx.doi.org/10.1038/nchembio755
    » http://dx.doi.org/10.1038/nchembio755
  • 87
    Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B, et al. Withaferin A targets heat shock protein 90 in pancreatic cancer cells. Biochem Pharmacol. 2010;79(4):542-51, http://dx.doi.org/10.1016/j.bcp.2009.09.017
    » http://dx.doi.org/10.1016/j.bcp.2009.09.017
  • 88
    Yang H, Shi G, Dou QP. The tumor proteasome is a primary target for the natural anticancer compound Withaferin A isolated from “Indian winter cherry”. Mol Pharmacol. 2007;71(2):426-37, http://dx.doi.org/10.1124/mol.106.030015
    » http://dx.doi.org/10.1124/mol.106.030015
  • 89
    Veras ML, Bezerra MZ, Braz-Filho R, Pessoa OD, Montenegro RC, do O Pessoa C, et al. Cytotoxic epimeric withaphysalins from leaves of Acnistus arborescens Planta Med. 2004;70(6):551-5, http://dx.doi.org/10.1055/s-2004-827156
    » http://dx.doi.org/10.1055/s-2004-827156
  • 90
    Veras ML, Bezerra MZ, Lemos TL, Uchoa DE, Braz-Filho R, Chai HB, et al. Cytotoxic withaphysalins from the leaves of Acnistus arborescens. J Nat Prod. 2004;67(4):710-3, http://dx.doi.org/10.1021/np0340347
    » http://dx.doi.org/10.1021/np0340347
  • 91
    Rocha DD, Balgi A, Maia AI, Pessoa OD, Silveira ER, Costa-Lotufo LV, et al. Cell cycle arrest through inhibition of tubulin polymerization by withaphysalin F, a bioactive compound isolated from Acnistus arborescens Invest New Drugs. 2012;30(3):959-66, http://dx.doi.org/10.1007/s10637-011-9649-x
    » http://dx.doi.org/10.1007/s10637-011-9649-x
  • 92
    Aoki T, Akashi T, Ayabe S. Flavonoids of leguminous plants: structure, biological activity, and biosynthesis. J Plant Res. 2000;113(4):475-88, http://dx.doi.org/10.1007/PL00013958
    » http://dx.doi.org/10.1007/PL00013958
  • 93
    Nakagawa M, Nakanishi K, Darko LL, Vick JA. Structures of cabenegrins AI and A-II, potent anti-snake venoms. Tetrahedron Lett. 1982;23(38):3855-8, http://dx.doi.org/10.1016/S0040-4039(00)87726-6
    » http://dx.doi.org/10.1016/S0040-4039(00)87726-6
  • 94
    Engler TA, Lynch KO, Reddy JP, Gregory GS. Synthetic pterocarpans with anti-HIV activity. Bioorg Med Chem Lett. 1993;3(6):1229-32, http://dx.doi.org/10.1016/S0960-894X(00)80321-2
    » http://dx.doi.org/10.1016/S0960-894X(00)80321-2
  • 95
    Salem MM, Werbovetz KA. Isoflavonoids and other compounds from Psorothamnus arborescens with antiprotozoal activities. J Nat Prod. 2006;69(1):43-9, http://dx.doi.org/10.1021/np0502600
    » http://dx.doi.org/10.1021/np0502600
  • 96
    Jiménez-González L, Álvarez-Corral M, Muãoz-Dorado M, Rodríguez-García I. Pterocarpans: interesting natural products with antifungal activity and other biological properties. Phytochem Rev. 2008;7(1):125-54.
  • 97
    Tesauro C, Fiorani P, D’Annessa I, Chillemi G, Turchi G, Desideri A, Erybraedin C. A natural compound from the plant Bituminaria bituminosa, inhibits both the cleavage and religation activities of human topoisomerase I. Biochem J. 2010;425(3):531-9, http://dx.doi.org/10.1042/BJ20091127
    » http://dx.doi.org/10.1042/BJ20091127
  • 98
    Goel A, Kumar A, Raghuvanshi A. Synthesis, stereochemistry, structural classification, and chemical reactivity of natural pterocarpans. Chem Rev. 2013;113(3):1614-40, http://dx.doi.org/10.1021/cr300219y
    » http://dx.doi.org/10.1021/cr300219y
  • 99
    Falcão MJ, Pouliquem YB, Lima MA, Gramosa NV, Costa-Lotufo LV, Militão GC, et al. Cytotoxic flavonoids from Platymiscium floribundum J Nat Prod. 2005;68(3):423-6, http://dx.doi.org/10.1021/np049854d
    » http://dx.doi.org/10.1021/np049854d
  • 100
    Militão GC, Bezerra DP, Pessoa C, de Moraes MO, da Ponte FA, Lima MA, et al. Comparative cytotoxicity of 2, 3, 9-trimethoxypterocarpan in leukemia cell lines (HL-60, Jurkat, Molt-4, and K562) and human peripheral blood mononuclear cells. J Nat Med. 2007;61(2):196-9, http://dx.doi.org/10.1007/s11418-006-0118-3
    » http://dx.doi.org/10.1007/s11418-006-0118-3
  • 101
    Militão GC, Dantas IN, Pessoa C, Falcão MJ, Silveira ER, Lima MA, et al. Induction of apoptosis by pterocarpans from Platymiscium floribundum in HL-60 human leukemia cells. Life Sci. 2006;78(20):2409-17, http://dx.doi.org/10.1016/j.lfs.2005.09.044
    » http://dx.doi.org/10.1016/j.lfs.2005.09.044
  • 102
    Militão GC, Prado MP, Pessoa C, de Moraes MO, Silveira ER, Lima MA, et al. Pterocarpans induce tumor cell death through persistent mitotic arrest during prometaphase. Biochimie. 2014;104:147-55, http://dx.doi.org/10.1016/j.biochi.2014.06.005
    » http://dx.doi.org/10.1016/j.biochi.2014.06.005
  • 103
    Klein E, DeBonis S, Thiede B, Skoufias DA, Kozielski F, Lebeau L. New chemical tools for investigating human mitotic kinesin Eg5. Bioorg Med Chem. 2007;15(19):6474-88, http://dx.doi.org/10.1016/j.bmc.2007.06.016
    » http://dx.doi.org/10.1016/j.bmc.2007.06.016
  • 104
    Müller C, Gross D, Sarli V, Gartner M, Giannis A, Bernhardt G, et al. Inhibitors of kinesin Eg5: antiproliferative activity of monastrol analogues against human glioblastoma cells. Cancer Chemother Pharmacol. 2007;59(2):157-64.
  • 105
    Kapoor TM, Mayer TU, Coughlin ML, Mitchison TJ. Probing spindle assembly mechanisms with monastrol, a small molecule inhibitor of the mitotic kinesin, Eg5. J Cell Biol. 2000;150(5):975-88, http://dx.doi.org/10.1083/jcb.150.5.975
    » http://dx.doi.org/10.1083/jcb.150.5.975
  • 106
    Gartner M, Sunder-Plassmann N, Seiler J, Utz M, Vernos I, Surrey T, et al. Development and biological evaluation of potent and specific inhibitors of mitotic Kinesin Eg5. ChemBioChem. 2005;6(7):1173-7, http://dx.doi.org/10.1002/cbic.200500005
    » http://dx.doi.org/10.1002/cbic.200500005
  • 107
    Weaver BA. How Taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25(18):2677-81, http://dx.doi.org/10.1091/mbc.e14-04-0916
    » http://dx.doi.org/10.1091/mbc.e14-04-0916
  • 108
    Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359-86, http://dx.doi.org/10.1002/ijc.29210
    » http://dx.doi.org/10.1002/ijc.29210
  • 109
    Shi YW, Yuan W, Wang X, Gong J, Zhu SX, Chai LL, et al. Combretastatin A-4 efficiently inhibits angiogenesis and induces neuronal apoptosis in zebrafish. Sci Rep. 2016;6(1):30189, http://dx.doi.org/10.1038/srep30189
    » http://dx.doi.org/10.1038/srep30189
  • 110
    Newman DJ, Cragg GM. Natural Products as Sources of New Drugs from 1981 to 2014. J Nat Prod. 2016;79(3):629-61, http://dx.doi.org/10.1021/acs.jnatprod.5b01055
    » http://dx.doi.org/10.1021/acs.jnatprod.5b01055
  • 111
    Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, et al. Antibody-Drug Conjugates for Cancer Therapy: Chemistry to Clinical Implications. Pharmaceuticals (Basel). 2018;11(2). pii: E32, http://dx.doi.org/10.3390/ph11020032
    » http://dx.doi.org/10.3390/ph11020032

Publication Dates

  • Publication in this collection
    2018

History

  • Received
    29 May 2018
  • Accepted
    9 Oct 2018
Creative Common - by 4.0
This is an Open Access article distributed under the terms of the Creative Commons License (https://creativecommons.org/licenses/by/4.0/) which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is properly cited.
Faculdade de Medicina / USP Rua Dr Ovídio Pires de Campos, 225 - 6 and., 05403-010 São Paulo SP - Brazil, Tel.: (55 11) 2661-6235 - São Paulo - SP - Brazil
E-mail: clinics@hc.fm.usp.br