Next Article in Journal
Molecular Pathogenesis of Fibrosis, Thrombosis and Surfactant Dysfunction in the Lungs of Severe COVID-19 Patients
Next Article in Special Issue
Nanobody-Based EGFR-Targeting Immunotoxins for Colorectal Cancer Treatment
Previous Article in Journal
Functional Resilience of Mutually Repressing Motifs Embedded in Larger Networks
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy

Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine
Biomolecules 2022, 12(12), 1843; https://doi.org/10.3390/biom12121843
Submission received: 29 October 2022 / Revised: 2 December 2022 / Accepted: 6 December 2022 / Published: 10 December 2022
(This article belongs to the Special Issue Novel Molecules for Cancer Treatment 2.0)

Abstract

:
Cancer accounts for numerous deaths each year, and it is one of the most common causes of death worldwide, despite many breakthroughs in the discovery of novel anticancer candidates. Each new year the FDA approves the use of new drugs for cancer treatments. In the last years, the biological targets of anticancer agents have started to be clearer and one of these main targets is tubulin protein; this protein plays an essential role in cell division, as well as in intracellular transportation. The inhibition of microtubule formation by targeting tubulin protein induces cell death by apoptosis. In the last years, numerous novel structures were designed and synthesized to target tubulin, and this can be achieved by inhibiting the polymerization or depolymerization of the microtubules. In this review article, recent novel compounds that have antiproliferation activities against a panel of cancer cell lines that target tubulin are explored in detail. This review article emphasizes the recent developments of tubulin inhibitors, with insights into their antiproliferative and anti-tubulin activities. A full literature review shows that tubulin inhibitors are associated with properties in the inhibition of cancer cell line viability, inducing apoptosis, and good binding interaction with the colchicine binding site of tubulin. Furthermore, some drugs, such as cabazitaxel and fosbretabulin, have been approved by FDA in the last three years as tubulin inhibitors. The design and development of efficient tubulin inhibitors is progressively becoming a credible solution in treating many species of cancers.

1. Introduction

Cancer accounts for numerous deaths each year, and it is one of the most common causes of death worldwide, despite many breakthroughs in the discovery of novel anticancer candidates [1,2]. Each new year, the FDA approves the use of new drugs for cancer treatments, but due to multiple drug resistance and serious side effects, current treatments become non-ideal therapy; because of that, great efforts to discover a new agent with fewer toxic effects are necessary [3,4,5]. Many new chemical structures were designed and synthesized regarding cancer’s biological targets, such as cyclin-dependent kinase (CDK), epidermal growth factor (EGF), Ras, and tubulin proteins. These targets were classified as the main targets of new anticancer candidates [6,7], and with regards to this, tubulin is considered as one of the most useful and strategic molecular targets for antitumor drugs [8]. Microtubules play an important role in intracellular cell division, as well as in transportation. Tubulin protein polymerizes into long chains, or filaments, to build hollow fibers, or microtubules. These fibers work like a skeletal system for living cells and are the clear target for anticancer agents [9,10]. The targeting of tubulin protein by the inhibition of microtubule formation usually induces apoptosis (programmed cell death) [11,12].
The design and discovery of new tubulin inhibitors (TIs) targeting the colchicine binding site appears an attractive path for improving and advancing tubulin inhibitors [13]. TIs are less prone to develop multi-drug resistance (MDR) in comparison with vinca alkaloids and taxanes because they are poor substrates for efflux mechanism P-gp [14,15]. Moreover, many TIs have disadvantages, such as serious side effects like neurotoxicity, and chemical instability [16]. Currently, an FDA-approved drug, fosbretabulin (combretastatin A-4 phosphate), which is utilized for the treatment of thyroid cancer, can specifically target the colchicine binding site of tubulin [17,18].
Therefore, TIs that bind to the colchicine site has received extraordinary attention in the last 10 years [19]. Based on this data, numerous microtubule targeting agents have been discovered as effective TIs for various cancer forms in the last decade, and some of these chemicals have entered clinical trials. This review aimed to describe recent advances in the development of chemical structures that target tubulin at the colchicine binding site as promising anticancer agents.

2. Combretastatin A-4 Analogues

Combretastatin A-4 (CA-4) (Figure 1) is the most common member of the combretastatin family, which was isolated from the African tree Combretum caffrum [20,21,22]. CA-4 exhibits strong antimitotic activity by binding to the colchicine binding site and entered phase II and phase III studies in clinical trials [23,24]. CA-4 has various pharmacokinetic disadvantages, such as poor water solubility [25,26,27], as well as having a short plasma half-life and instability due to isomerization from active cis isomer to inactive trans isomer under in vivo conditions [28,29]. To improve the low water solubility of CA-4, researchers developed CA-4, and optimized this pharmacokinetic challenge by innovating CA-4P (fosbretabulin) (Figure 1), which was applied for thyroid cancer, and was approved by the FDA in 2018 [17,30]. Moreover, many fosbretabulin salts were developed as fosbretabulin disodium and fosbretabulin tromethamine, as well as new derivatives being discovered, such as Oxi4503 and Ombrabulin (Figure 1). This has been trialed as monotherapy, as well as in combination with well-known anticancer agents such as cisplatin, paclitaxel, carboplatin, pazopanib, and bevacizumab [31,32]. Recently, several studies have been performed on the CA-4 derivatives and their antiproliferative activities targeting tubulin were investigated [33,34,35,36,37].
Researchers attempted to design and synthesize analogs of CA-4 by replacing the linker with hetero atoms (Si, Se, and N). Compound st.1 (Figure 2) replaced the double bond between the two phenyl rings of CA-4 (linker) with a silicone atom; they were trying to design a compound that had a linker with a similar distance between two phenyl rings compared to CA-4. As a result, st.1 showed inhibition of tubulin polymerization at 30 μM concentration, as well as the antiproliferative activities against breast cancer cell line (MCF7), with an IC50 value 7 nM [38]. In another study, the linker of CA-4 was replaced with selenide, and st.2 (Figure 2) was active at nM concentration against MCF-7 cancer cell lines. This structure also showed potent activities as a tubulin polymerization inhibitor more active than CA-4 itself [39]. Soussi et al. synthesized CA-4 analogs methylated amine instead of CA-4 linker, and compound st.3 (Figure 2) showed excellent anticancer activity at an average nanomolar level of mean GI50 values and inhibited tubulin assembly at a micromolar level. In addition, this compound, showed cell cycle arrest in the G2/M phase and induced apoptosis at very low concentration [40].
In various studies, researchers have placed heterocycle (pyrazole, isoxazole, tetrazole, thiazole, imidazole, pyrrole, oxazole, and β-lactam) instead of the double bound of CA-4 as a linker between phenyl rings. The incorporation of these heterocycles was very important for improving the water solubility of the CA-4 analogs [41,42,43,44,45,46]. Some introduced the pyrazole and tetrazole rings (St.4 and St.5; Figure 3) instead of the double bond of CA-4; both of these structures showed tubulin polymerization inhibition with IC50 values 3 and 2 μM, respectively [34,47]. Wang et al. replaced the linker with an imidazole ring, compound St.6 (Figure 3), with potent antiproliferative activities, and its pharmacokinetic properties were also perfect with 82% bioavailability in rats [48]. In another work, the thiazole ring was used as a linker, and it was substituted with methylamine. In terms of the structure-activity relationship, it was clear that NHCH3 substituent at the fifth position of the thiazole ring showed better antiproliferative activities than methyl or dimethylamine. However, in this series, compound St.7 (Figure 3) was one of the most potent compounds against MCF-7 cancer cell lines with IC50 values in the nanomolar level, and it showed the inhibition of tubulin polymerization with IC50 1.3 μM in comparison with CA-4 IC50 1.2 μM [49].
Several researchers have tried to use four-membered ring β-lactam as a bridge in the CA-4; these derivatives showed potent antiproliferative and antimitotic activities [50,51,52,53], and compound St.8 (Figure 4) showed significant cytotoxicity against MCF7 cancer cell lines with IC50 values in nanomolar with significant in vitro inhibition of tubulin polymerization [54]. On the same core cycle, another group synthesized a series of (3-substituted 1,4-diaryl-2-azetidine) and compound St.9 (Figure 4) was significantly the cell proliferation in IC50 range 31-63 nM, as well as its tubulin polymerization inhibition’s IC50 was around 3.5 µM. According to X-ray crystallography, this compound was binding to the colchicine binding site in tubulin in a similar mode like that of colchicine [55].
Seven membered rings as linkers were conducted in the synthesis of novel CA-4 derivatives, and one of these researches innovated a compound St.10 (Figure 4), which inhibits the tubulin polymerization by binding to the colchicine binding site of tubulin. This compound showed potent antiproliferative activities against various kinds of human cancer cell lines, with IC50 values in nanomolar level, as well as the flow cytometric analysis results showing that this structure can induce cell cycle arrest in G2/M phase and apoptosis in A549 cancer cell line [56]. Another study was conducted on these basic core structures; a series of novel benzoxepins was synthesized and evaluated as anticancer agents, and among this series the most potent compound was St.11 (Figure 4). This displayed antiproliferative activity with IC50 range 1.5-8 nM against different kinds of cancer cell lines (HCT116, K562, H1299, and MDA-MB231) and it also inhibited the tubulin polymerization at a micromolar range (IC50 = 3.8 µM) [57].
Recent reports indicate that derivatives of pyrazole, isoxazole, and phenyl cinnamide, play an important role in the development of potential cytotoxic agents. In many kinds of research, the linker of CA-4 was replaced with amide or heterocyclic amide, and these derivatives showed significant antimitotic activities [58,59,60]. Phenyl cinnamides compound St.12 (Figure 5) was shown to bind to tubulin, causing the inhibition of tubulin polymerization [61]. A series of pyrazole or isoxazole-linked arylcinnamides were designed and synthesized as anticancer agents; these compounds showed moderate to potent antiproliferative activities against HeLa, DU-145, A549, and MDA-MB231 cancer cell lines. In addition, among these structures, compound St.13 (Figure 5) significantly depolymerizes tubulin with an IC50 value of about 1.5 μM. This compound was found to arrest the cells cycles in G2/M phase [62].

3. Indole Analogues

Many natural and synthetic compounds containing an indole ring showed various pharmacological activities, mainly anticancer properties, and many of these compounds target the tubulin protein to combat cancer cell proliferation [63,64,65].
The indole ring is considered a commonly distributed heterocycle in nature, and synthetic compounds, and it has been becoming an essential core structure in many pharmaceutical compounds. It possesses various pharmacological activities, such as antifungal, antioxidant [66], antiviral [67,68], antidepressant, anticonvulsant, and anti-inflammatory [65], and recently was involved in the discovery of new anticancer agents [69,70,71,72,73,74] as well as tubulin inhibitors [33,75,76]. Arylthioindole derivatives were one of the main classes of indole-containing compounds with tubulin inhibition activities. This family mainly contained trimethoxyphenyl moiety, beside the indole core structure [77]. St.14, St.15, St.16, and St.17 (Figure 6) were potent tubulin inhibitors with IC50 values of 1.6, 2, 0.99, and 0.67 μM, respectively. They also exhibited antiproliferative activities with IC50 in nanomolar level against various cancer cell lines, such as MCF-7 and U937 [74]. Many structures were developed and discovered with various substitutes on the indole ring, and the trimethoxyphenyl moiety remained because of its supposed interactions with tubulin amino acids. One of these structures, St.18 (Figure 6), exhibits potent antiproliferative activity with IC50 values in the nanomolar level; it also showed disruption of the microtubule network [78]. St.19 (Figure 6) showed potent activity in various cancer cell lines without effects on normal cell lines [79]. Bis-indole derivatives were developed as antimitotic agents [80] and St.20 (Figure 6) showed an inhibition effect on tubulin polymerization with an IC50 value of 7.5 µM, in addition to displaying anti-proliferative activity against the A549 cancer cell line with an IC50 value of 2 μM [81].
Aroylindoles have also been considered as one of the major classes of indole-containing compounds with tubulin-inhibiting effects [82]. St.21 (Figure 7) was developed as a tubulin inhibitor agent and it interfered with the colchicine binding site of tubulin [83]. Additionally, St.22 (Figure 7) is an indolyl-phenylmethanone derivative and showed potent antimitotic effects in human cancer cell lines, as well as potent antiproliferative activities against various kinds of cancer cell lines such as glioblastoma, breast, and gastric cancer cells [84]. In another study, aroylindole derivatives were discovered and synthesized as potent antitumor and antimitotic agents, St.23 and St.24 (Figure 7), with OH and NH2 at the 4th position of indole showing potent anti-tubulin activity with IC50 values of 0.6 and 0.9 μM, respectively. They also showed antiproliferative activity with IC50 in nanomolar level against different kinds of cancer cell lines [64,85].
Many kinds of research have been focused to discover hybrid chemical agents with antimitotic activities [86], including indole-heterocycles hybrids derivatives with promising anticancer activities [87,88]. Pyrazole-oxindole derivatives were also developed and evaluated on tubulin polymerization, and different kinds of cancer cell lines such as HeLa, A549, and MCF7, St.25, St.26, and St.27 (Figure 7), exhibited anti-tubulin activities with IC50 values in the range 5.90–9.20 μM [89]. Meanwhile, St.28 (Figure 7) with indole-amino-pyrazolyl derivatives was synthesized and inhibited tubulin polymerization with IC50 values of 0.28 μM [90]. In new research on a indole-furanone hybrid, derivatives were synthesized and developed as anti-tubulin derivatives; the most potent compound, St.29 (Figure 7), has anticancer and antimitotic potency at the micromolar level [91]. In another recent work, the authors developed and synthesized an indole-heterocycle hybrid like the furan ring, and these structures showed potent antiproliferative activities. Among the synthesized compounds, St.30 (Figure 7) was able to induce cell cycle arrest at the G2/M phase on A549 cancer cell line. Additionally, this structure also exhibited tubulin polymerization inhibitory activities, whereas when the bromine substitution was added to indole ring and methoxy on furan ring (St.31; Figure 7) and the anticancer activities were better than St.30 with IC50 value under 0.5 μM against HuCCA-1 and HepG2 cancer cell lines [92].
The indole-amide class was considered one of the main classes of indole derivatives with anticancer and/or anti-tubulin activities [22,91,93]. A series of indole-acrylamide was synthesized and developed as anti-tubulin inhibitors, and St.32 (Figure 7) showed inhibitory activities of tubulin polymerization. As well as this compound causing cell cycle arrest at the G2/M phase of HeLa and HL-60 cancer cell lines, it also induced apoptosis by the activation of caspase-3 [22]. The same group of researchers tried to target the tubulin by adding various substituents, such as methyl, nitrile, carboxylic acid, and ester, to the linker of St.32. However, the derivative with the nitrile group was considered as a tubulin polymerization inhibitor [94]. Another novel series of indole-based oxalamide was developed and designed, amongst which St.33 (Figure 7) exhibits potent anticancer activities against HeLa, PC-3, and HCT-116 cancer cell lines. Meanwhile, the immunocytochemistry observed a significant loss of microtubule contents after the treatment of the cells with the compound, and confirmed the inhibition of tubulin polymerization accordingly [95].

4. Thiophene and Quinolone Analogs

Compounds containing thiophene rings have different biological activities, such as anticancer and anti-tubulin. A great deal of work has reported that the thiophene nucleus is an important structural heterocycle in antimitotic compounds [96,97,98]. A series of thiophene derivatives were designed, synthesized, and evaluated for anticancer activity against various cancer cell lines, as well as showing the inhibition of tubulin polymerization. Among the synthesized series both compounds St.34 and St.35, (Figure 8), showed IC50 values of less than 1 nM against HeLa, HL-40, MCF-7, and HT-29 cancer cell lines, while the IC50 of tubulin polymerization was 0.88 and 0.70 μM, respectively [99]. In another study, 15 novel compounds were synthesized and evaluated against 60 kinds of cancer and normal cell lines, and the most potent compound was St.36 (Figure 8), with benzothiophene moiety; IG50 values of this compound were less than 10 nM against most of tested cancer cell lines, and the IC50 value towards the inhibition of tubulin polymerization was 1.7 µM [100].
In a recent study, a series of tetrahydrobenzo[b]thiophene derivatives were synthesized and evaluated against colorectal cancer. The most active compound, St.37 (Figure 8), showed moderate antiproliferative activities with IC50 values of 81.50 and 71.00 μg/mL against LoVo cells and HCT-116 cells cell lines. Meanwhile, molecular docking analysis in the colchicine binding site supposed good binding affinity of this compound [101]. Another new novel series of thiophene derivatives were synthesized and evaluated as anticancer agents and some of these compounds were considered significant apoptosis-inducing compounds. Compounds St.38 and St.39 (Figure 8) showed the greatest anticancer activity against HeLa and HT-29 cancer cell lines, with IC50 values of 0.06–0.50 μM. The colchicine binding studies were conducted to evaluate the tubulin polymerization inhibition, and the most active compound among the synthesized series was St.39, with a moderate inhibition percentage (30%) at 5 μM concentration in comparison with CA-4, with a potent inhibition percentage (97%) of the binding to [3H]colchicine to tubulin [102]. Another series of tetrahydrothiophene were designed, synthesized, and evaluated for anticancer activities against cancer cell lines and the inhibition of tubulin polymerization, as well as the effect on cell cycle phases. Compounds St.40 and St.41, (Figure 8), with trimethoxyanilino and nitrile groups as important groups for the activities, these two compounds exhibited significant antiproliferative activities against L1210, CEM, and HeLa cancer cell lines with IC50 values in range 1.10–4.70 µM. They showed cell cycle arrest and accumulation at the G2/M phase and their IC50 values regarding the inhibition of tubulin polymerization were 3.8 and 3.4 µM, respectively, compared to the CA-4 IC50 value of 0.54 µM [103].
Quinoline heterocycle considers one of the important heterocycles, which possess various biological activities including anticancer, antimicrobial, anti-HIV, and anti-inflammatory [104]. In the last years, new chemical series containing quinoline and quinoline isosteres were designed to have antiproliferative activities [105,106] and target microtubules [107,108,109].
A series of quinoline-indole derivatives were designed and synthesized to target tubulin and inhibit its functions, this series was a CA-4 analog. Additionally, the main core structure is similar to St.18 (Figure 6); two compounds, St.42 and St.43 (Figure 9), showed the most potent antiproliferative against various cancer cell lines (HepG2, KB, HCT-8, MDA-MB-231, and H22), with IC50 values < 10 nM. Both of these compounds (St.42 and St.43) also effectively inhibited the tubulin polymerization with IC50 values 2.54 and 2.09 μM, respectively, in comparison with CA-4 IC50 2.12 μM [108]. In recent work, researchers innovated a quinoline series by using the three-dimensional quantitative SAR strategy, and these series were virtually designed and evaluated as new anticancer/tubulin inhibitor agents. St.44 (Figure 9) was one of the most active ligands regarding its possible binding interactions with the colchicine binding site [110].
Another series of quinoline derivatives were developed, synthesized, and evaluated as anticancer/tubulin inhibitors, amongst the synthesized series. St.45 (Figure 9) was the most potent compound against HepG-2, B16-F1, HeLa, and MCF-7 cancer cell lines with IC50 value ranging from 0.261–2.047 μM. Furthermore, this compound has significant inhibition towards tubulin polymerization with IC50 12.38 μM in comparison with CA-4 IC50 1.84 μM [111]. In new work, cyano-pyrrolo-quinoline derivatives were synthesized and evaluated for their antiproliferative activity against 60 human cancer cell lines, and the most active compound, St.46 (Figure 9), has GI50 values > 2.0 μM against most of the tested cancer cell lines. Additionally, in vitro assays and molecular docking studies regarding this compound found a significant binding interaction with tubulin [112]. In a recently published work, a new novel series of quinoline CA-4-based analogs were designed and evaluated as anticancer agents. One of the synthesized compounds, St.47 (Figure 9), was considered a tubulin polymerization inhibitor by the performed mechanistic studies, and regarding cell cycle analysis it was made in accumulation and arrest in G2/M phase. In addition, this compound was the most active compound against MCF-7, HL-60, HCT-116, and HeLa cancer cell lines with IC50 values < 42 nM [113]. In a series of quinoline-pyrazole and quinoline-pyridone derivatives, St.48 and St.49 (Figure 9) showed the most potent tubulin polymerization inhibitory activities with IC50 values of 9.11 and 10.5 nM, respectively. These two structures showed significant antiproliferative activities against MCF-7, HepG-2, and HCT-116 cancer cell lines [114].

5. Chalcone Analogs

Chalcones (1,3-diaryl-2-proper-1-ones) are naturally occurring precursors of flavonoids, and these compounds have broad pharmacological activities including anti-cancer, antifungal, anti-inflammatory, and antioxidative activity [97,115,116,117,118]. A novel series of imidazole-chalcone derivatives were developed, synthesized, and evaluated as tubulin inhibitors. The most potent anticancer agent amongst the developed derivatives was St.50 (Figure 10), A549, and MCF-7 cancer cell line with IC50 values of 7.05 and 9.88 μM, respectively. However, this compound also inhibited tubulin polymerization in a similar mode to CA-4 [119]. In another study, diaryl chalcone derivates were synthesized, and compound St.51 (Figure 10) was the most potent anticancer agent against HCT116, HepG2, and MCF-7 cancer cell lines, with IC50 values < 6.31 μM. Additionally, the tubulin polymerization assay and molecular docking analysis supposed that this compound could effectively inhibit tubulin polymerization, and bind very well in colchicine binding site [120]. In another work, triazolo-quinoxaline chalcone derivatives were developed and two compounds, St.52 and St.53 (Figure 10), exhibited potent antiproliferative activities against MCF-7, HCT-116 and HepG2 cancer cell lines with IC50 range 0.84-15.4 μM. They could inhibit the EGFR with IC50 values of 39 and 83 nM, respectively; additionally they inhibit the tubulin polymerization with IC50 values 8.84 and 14.7 μM [109].
In another study, a series of naphthalene-chalcone derivatives were developed, synthesized, and evaluated as anticancer agents. Almost all of the synthesized derivatives showed considerable anticancer activities against the MCF-7 cancer cell line, and the most potent compound was St.54 (Figure 10), with an IC50 value of 1.42 µM, with lower cytotoxicity against normal cell line Hek293t. Additionally, this compound inhibits the tubulin polymerization with IC50 value around 8 µM, in comparison with colchicine IC50 value (10.6 µM) [121]. By developing St.54, a series of naphthalene-thiazole derivatives were designed and among the synthesized compounds, St.55 (Figure 10) was the most potent compound against MCF-7 and A549 cancer cell lines with IC50 values > 1 μM. It was found significant to inhibit the tubulin polymerization with IC50 value 3.3 µM [122].

6. Trimethoxy Phenyl Analogs

In the last decade, many trimethoxyphenyl-based structures were designed, developed, and synthesized as promising anticancer agents that could target tubulin protein, and some of these compounds reached clinical trials, or were approved by the FDA for cancer treatment [123,124,125]. Starting with CA-4P (Figure 1), which was approved for thyroid cancer, many other trimethoxyphenyl-based compounds entered the clinical trials for specific cancer types including St.56 (BNC-105p; Figure 11), which was found to have considerable potency and an inhibitory effect against the growth of different kinds of cancer cell lines with a broader therapeutic index than CA-4P in vivo. It entered phase I of metastatic renal cell carcinoma malignant [126].
However, a lot of work with structures containing trimethoxyphenyl moiety were mentioned in the previous sections, and various structures with this moiety were designed and synthesized as promising anticancer agents. A series of isoxazole-carboxamide derivatives, such as St.57 (Figure 11), exhibited potent antiproliferation activities against a panel of cancer cell lines; potent anticancer activities of St.57 was observed against Huh7, MCF7, and HCT116, with IC50 values 0.7, 3.6, and 1.3 μM, respectively, as well as potent activities against another hepatocellular carcinoma cell lines such as HepG2, Mahlavu, and SNU475 cancer (IC50 < 3.1 μM) [127]. Recently, many works focused on this moiety, and in a series of trimethoxyphenyl-pyrazolo-amine, St.58 (Figure 11) was the most potent compound. The free amine group was supposed to play an essential role in the antiproliferative effects, and this compound exhibited significant anticancer activities against MCF-7, HCT-116, and HeLa cancer cell lines with IC50 values < 0.26 μM. Additionally, the mechanistic studies showed considerable inhibition towards tubulin polymerization activity with IC50 value of 14 μM [128]. In another recent study, the trimethoxyphenyl moiety with benzimidazole was used as CA-4 based structure, and compound St.59 (Figure 11) exhibited the most potent effects against MCF-7, SGC-7901, and A549 cancer cell lines with IC50 values < 0.20 μM. At the same time, this compound inhibited tubulin polymerization by disrupting the cell microtubule networks, and cell cycle arrests were observed in G2/M phase. Regarding the in vivo study, this compound exhibited potent antitumor efficacy [129]. In a similar study to the previous one, researchers changed the benzimidazole of St.59 with triazolo-pyrimidine, and in this series St.60 (Figure 11) showed potent antiproliferative activities against a panel of cancer cell lines and specifically against HeLa cancer cell line with IC50 value of 0.06 μM and cell cycle arrest was observed in the G2/M phase. However, this compound inhibited tubulin polymerization with IC50 1.3 μM, which was better than the positive control CA-4 IC50 value (4.22 μM) [130].
In another study with trimethoxyphenyl moiety, a series of the seven-membered ring (benzothiazepine), which is similar to CA-4 analogs (St.10 and St.11; Figure 4), and compound St.61 (Figure 11) was the most potent compound among this series with significant antiproliferative activities against MCF-7, HeLa, Ht29, and A549 cancer cell lines. The IC50 values were < 2 μM, as well as this compound inhibiting tubulin polymerization with IC50 values of 1.20 μM [131]. A series of thiazole-thiones containing trimethoxyphenyl moiety was designed and developed recently as anticancer agents. Among the synthesized derivatives, compounds St.62 and St.63 (Figure 11) were the most potent structures against the MCF-7 cancer cell line with IC50 values of 1.14 and 2.41 μg/mL, respectively. Regarding the obtained results of tubulin polymerization inhibition, St.62 was more potent than St.63 with IC50 values 5.14 and 9.97 μg/mL, respectively [132]. In the newest work, a series of indole-acrylamide were developed and synthesized as promising antimitotic agents, with the antiproliferation activities of the synthesized compounds against a panel of cancer cell lines, and particularly focusing on hepatocellular carcinoma. Among this series, compound St.64 (Figure 11) exhibited potent antiproliferative activities. Additionally, this compound, was determined to be a tubulin polymerization inhibitor with IC50 18 μM. Furthermore, cell cycle arrest was observed in the G2/M phase in the Huh7 cancer cell line [94].

7. Approved and Promising Antimitotic Agents

In 1963, the first tubulin targeting drug (vincristine) was approved by the FDA for the treatment of cancer. Researchers began to take an interest in this class and many structures and drugs were discovered and approved for this purpose [133]. However, in the last decades, many TI agents have entered clinical trials and shown promising anticancer activities and some of these agents were approved to be used for certain kinds of serious cancers. Table 1 shows a list of drugs in clinical developments beside approved drugs that targeted tubulin as an anticancer agent [134].

8. Conclusions

In the last decades, great efforts have been made to discover tubulin inhibitors, and a few drugs have been approved by the FDA for the treatment of cancer by targeting tubulin as a molecular target. Unfortunately, most of the approved drugs for this target were associated with disadvantages, including low potency, drug resistance, and/or toxicity. However, because of these reasons, researchers are continuously attempting to develop and discover agents with ideal properties. Several groups were reached in this review article, and it was clear that the most important groups were CA-4 analogs, trimethoxyphenyl, and indole derivatives, and they exhibited potent antiproliferation and anti-tubulin activities. Various compounds with anti-tubulin activities were synthesized and developed, and it was clear that the compounds, which are similar to the CA-4 core structure and contain trimethoxyphenyl and indole, have very significant and considerable activities. In summary, this study focused on recent tubulin inhibitors, and the development of compounds with better selectivity, potency, and pharmacokinetic characteristics, will perhaps continue to receive fundamental attention in the following years, the results of which will change the perception of cancer treatment.

Funding

This research received no external funding.

Institutional Review Board Statement

The study did not require ethical approval for studies not involving humans or animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The author wishes to thank An-Najah National University for their support to carry out this work.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Boyle, P.; Levin, B. World Cancer Report 2008; WHO report; The International Agency for Research on Cancer: Lyon, France, 2008; pp. 12–510. [Google Scholar]
  3. Heptinstall, A.B.; Adiyasa, I.; Cano, C.; Hardcastle, I.R. Recent advances in CDK inhibitors for cancer therapy. Future Med. Chem. 2018, 10, 1369–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Baytas, S.N.; Inceler, N.; Yılmaz, A. Synthesis, cytotoxicity, and molecular properties prediction of novel 1,3-diarylpyrazole derivatives. Med. Chem. Res. 2013, 22, 4893–4908. [Google Scholar] [CrossRef]
  5. Hawash, M.; Jaradat, N.; Eid, A.M.; Abubaker, A.; Mufleh, O.; Al-Hroub, Q.; Sobuh, S. Synthesis of novel isoxazole–carboxamide derivatives as promising agents for melanoma and targeted nano-emulgel conjugate for improved cellular permeability. BMC Chem. 2022, 16, 47. [Google Scholar] [CrossRef] [PubMed]
  6. Patrick, G.L. An Introduction to Medicinal Chemistry; Oxford University Press: Oxford, UK, 2013. [Google Scholar]
  7. Hawash, M. Highlights on Specific Biological Targets; Cyclin-Dependent Kinases, Epidermal Growth Factor Receptors, Ras Protein, and Cancer Stem Cells in Anticancer Drug Development. Drug Res. 2019, 69, 471–478. [Google Scholar] [CrossRef]
  8. Singh, H.; Kumar, M.; Nepali, K.; Gupta, M.K.; Saxena, A.K.; Sharma, S.; Bedi, P.M.S. Triazole tethered C5-curcuminoid-coumarin based molecular hybrids as novel antitubulin agents: Design, synthesis, biological investigation and docking studies. Eur. J. Med. Chem. 2016, 116, 102–115. [Google Scholar] [CrossRef]
  9. Bhattacharyya, B.; Panda, D.; Gupta, S.; Banerjee, M. Anti-mitotic activity of colchicine and the structural basis for its interaction with tubulin. Med. Res. Rev. 2008, 28, 155–183. [Google Scholar] [CrossRef]
  10. Ducki, S. Antimitotic Chalcones and Related Compounds as Inhibitors of Tubulin Assembly. Anti-Cancer Agents Med. Chem. 2009, 9, 336. [Google Scholar] [CrossRef]
  11. Flynn, B.L.; Hamel, E.; Jung, M.K. One-Pot Synthesis of Benzo[b]furan and Indole Inhibitors of Tubulin Polymerization. J. Med. Chem. 2002, 45, 2670–2673. [Google Scholar] [CrossRef]
  12. Hamel, E. Evaluation of Antimitotic Agents by Quantitative Comparisons of Their Effects on the Polymerization of Purified Tubulin. Cell Biochem. Biophys. 2003, 38, 1–21. [Google Scholar] [CrossRef]
  13. Arnst, K.E.; Wang, Y.; Hwang, D.-J.; Xue, Y.; Costello, T.; Hamilton, D.; Chen, Q.; Yang, J.; Park, F.; Dalton, J.T. A potent, metabolically stable tubulin inhibitor targets the colchicine binding site and overcomes taxane resistance. Cancer Res. 2018, 78, 265–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Brossi, A.; Yeh, H.J.; Chrzanowska, M.; Wolff, J.; Hamel, E.; Lin, C.M.; Quin, F.; Suffness, M.; Silverton, J. Colchicine and its analogues: Recent findings. Med. Res. Rev. 1988, 8, 77–94. [Google Scholar] [CrossRef] [PubMed]
  15. Ben-Chetrit, E.; Levy, M. Colchicine: 1998 update. In Seminars in Arthritis and Rheumatism; Elsevier: Amsterdam, The Netherlands, 1998; pp. 48–59. [Google Scholar]
  16. Lu, Y.; Chen, J.; Wang, J.; Li, C.-M.; Ahn, S.; Barrett, C.M.; Dalton, J.T.; Li, W.; Miller, D.D. Design, synthesis, and biological evaluation of stable colchicine binding site tubulin inhibitors as potential anticancer agents. J. Med. Chem. 2014, 57, 7355–7366. [Google Scholar] [CrossRef] [Green Version]
  17. Abma, E.; Daminet, S.; Smets, P.; Ni, Y.; De Rooster, H. Combretastatin A4-phosphate and its potential in veterinary oncology: A review. Vet. Comp. Oncol. 2017, 15, 184–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Hawash, M.; Jaradat, N.; Abualhasan, M.; Amer, J.; Levent, S.; Issa, S.; Ibrahim, S.; Ayaseh, A.; Shtayeh, T.; Mousa, A. Synthesis, chemo-informatics, and anticancer evaluation of fluorophenyl-isoxazole derivatives. Open Chem. 2021, 19, 855–863. [Google Scholar] [CrossRef]
  19. Nepali, K.; Ojha, R.; Sharma, S.; MS Bedi, P.; L Dhar, K. Tubulin inhibitors: A patent survey. Recent Pat. Anti-Cancer Drug Discov. 2014, 9, 176–220. [Google Scholar] [CrossRef]
  20. Ley, C.D.; Horsman, M.R.; Paul, E.G.; Kristjansen, P.E.G. Early Effects of Combretastatin-A4 Disodium Phosphate on Tumor Perfusion and Interstitial Fluid Pressure. Neoplasia 2007, 9, 108–112. [Google Scholar] [CrossRef] [Green Version]
  21. Gutiérrez, S.T.; Oltra, S.D.; Falomir, E.; Murga, J.; Carda, M.; Marco, J.A. Synthesis of combretastatin A-4 O-alkyl derivatives and evaluation of their cytotoxic, antiangiogenic and antitelomerase activity. Bioorganic Med. Chem. J. 2013, 21, 7267–7274. [Google Scholar] [CrossRef] [Green Version]
  22. Baytas, S.N.; Inceler, N.; Yılmaz, A.; Olgac, A.; Menevse, S.; Banoglu, E.; Hamel, E.; Bortolozzi, R.; Viola, G. Synthesis, biological evaluation and molecular docking studies of trans-indole-3-acrylamide derivatives, a new class of tubulin polymerization inhibitors. Bioorganic Med. Chem. 2014, 22, 3096–3104. [Google Scholar] [CrossRef] [Green Version]
  23. Gao, M.; Zhang, D.; Jin, Q.; Jiang, C.; Wang, C.; Li, J.; Peng, F.; Huang, D.; Zhang, J.; Song, S. Combretastatin-A4 phosphate improves the distribution and antitumor efficacy of albumin-bound paclitaxel in W256 breast carcinoma model. Oncotarget Impact J. 2016, 7, 58133–58141. [Google Scholar] [CrossRef]
  24. Nathan, P.; Zweifel, M.; Padhani, A.R.; Koh, D.M.; Ng, M.; Collins, D.J.; Harris, A.; Carden, C.; Smythe, J.; Fisher, N.; et al. Phase I Trial of Combretastatin A4 Phosphate (CA4P) in Combination with Bevacizumab in Patients with Advanced Cancer. Clin. Cancer Res. 2012, 18, 3428–3439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Simoni, D.; Romagnoli, R.; Baruchello, R.; Rondanin, R.; Rizzi, M.; Pavani, M.G.; Alloatti, D.; Giannini, G.; Marcellini, M.; Riccioni, T. Novel combretastatin analogues endowed with antitumor activity. J. Med. Chem. 2006, 49, 3143–3152. [Google Scholar] [CrossRef] [PubMed]
  26. Stevenson, J.P.; Rosen, M.; Sun, W.; Gallagher, M.; Haller, D.G.; Vaughn, D.; Giantonio, B.; Zimmer, R.; Petros, W.P.; Stratford, M. Phase I trial of the antivascular agent combretastatin A4 phosphate on a 5-day schedule to patients with cancer: Magnetic resonance imaging evidence for altered tumor blood flow. J. Clin. Oncol. 2003, 21, 4428–4438. [Google Scholar] [CrossRef] [PubMed]
  27. Hawash, M.; Jaradat, N.; Abualhasan, M.; Qneibi, M.; Rifai, H.; Saqfelhait, T.; Shqirat, Y.; Nazal, A.; Omarya, S.; Ibrahim, T.; et al. Evaluation of cytotoxic, COX inhibitory, and antimicrobial activities of novel isoxazole-carboxamide derivatives. Lett. Drug Des. Discov. 2022, 19. [Google Scholar] [CrossRef]
  28. Tron, G.C.; Pirali, T.; Sorba, G.; Pagliai, F.; Busacca, S.; Genazzani, A.A. Medicinal chemistry of combretastatin A4: Present and future directions. J. Med. Chem. 2006, 49, 3033–3044. [Google Scholar] [CrossRef]
  29. Aprile, S.; Del Grosso, E.; Tron, G.C.; Grosa, G. In vitro metabolism study of combretastatin A-4 in rat and human liver microsomes. Drug Metab. Dispos. 2007, 35, 2252–2261. [Google Scholar] [CrossRef] [Green Version]
  30. Eid, A.M.; Hawash, M.; Amer, J.; Jarrar, A.; Qadri, S.; Alnimer, I.; Sharaf, A.; Zalmoot, R.; Hammoudie, O.; Hameedi, S. Synthesis and Biological Evaluation of Novel Isoxazole-Amide Analogues as Anticancer and Antioxidant Agents. BioMed Res. Int. 2021, 2021, 6633297. [Google Scholar] [CrossRef]
  31. Li, D.-D.; Qin, Y.-J.; Zhang, X.; Yin, Y.; Zhu, H.-L.; Zhao, L.-G. Combined Molecular Docking, 3D-QSAR, and Pharmacophore Model: Design of Novel Tubulin Polymerization Inhibitors by Binding to Colchicine-binding Site. Chem. Biol. Drug Des. 2015, 86, 731–745. [Google Scholar] [CrossRef]
  32. Reddy, N.R.; Gouse, S.; Selvaraju, S.; Baskaran, S. Domino Semipinacol/Iterative Aldol/Iso-Nazarov Cyclization to Triaryl-cyclopentenone: Enantioselective Synthesis of Combretastatin A-4 Analogues. Org. Lett. 2022, 24, 4240–4245. [Google Scholar] [CrossRef]
  33. Reddy, T.S.; Rai, S.; Koppula, S.K. Synthesis of indole-tetrazole coupled aromatic amides; In vitro anticancer activity, in vitro tubulin polymerization inhibition assay and in silico studies. J. Mol. Struct. 2022, 1267, 133556. [Google Scholar] [CrossRef]
  34. Ohsumi, K.; Hatanaka, T.; Fujita, K.; Nakagawa, R.; Fukuda, Y.; Nihei, Y.; Suga, Y.; Morinaga, Y.; Akiyama, Y.; Tsuji, T. Syntheses and antitumor activity of cis-restricted combretastatins: 5-membered heterocyclic analogues. Bioorganic Med. Chem. Lett. 1998, 8, 3153–3158. [Google Scholar] [CrossRef]
  35. Yang, T.; Wang, Y.; Li, Z.; Dai, W.; Yin, J.; Liang, L.; Ying, X.; Zhou, S.; Wang, J.; Zhang, X.; et al. Targeted delivery of a combination therapy consisting of combretastatin A4 and low-dose doxorubicin against tumor neovasculature. Nanomedicine 2012, 8, 81–92. [Google Scholar] [CrossRef] [PubMed]
  36. Huang, X.; Chen, Y.; Zhong, W.; Liu, Z.; Zhang, H.; Zhang, B.; Wang, H. Novel combretastatin A-4 derivative containing aminophosphonates as dual inhibitors of tubulin and matrix metalloproteinases for lung cancer treatment. Eur. J. Med. Chem. 2022, 244, 114817. [Google Scholar] [CrossRef] [PubMed]
  37. Mustafa, M.; A Mostafa, Y.; E Abd Elbaky, A.; Mohamed, M.; Abdelhamid, D.; MN Abdelhafez, E.; Aly, O.M. Combretastatin A-4 analogs: Past, present, and future directions. Octahedron Drug Res. 2022, 1, 55–64. [Google Scholar] [CrossRef]
  38. Nakamura, M.; Kajita, D.; Matsumoto, Y.; Hashimoto, Y. Design and synthesis of silicon-containing tubulin polymerization inhibitors: Replacement of the ethylene moiety of combretastatin A-4 with a silicon linker. Bioorganic Med. Chem. J. 2013, 21, 7381–7391. [Google Scholar] [CrossRef] [PubMed]
  39. dos Santos, E.d.A.; Hamel, E.; Bai, R.; Burnett, J.C.; Tozatti, C.S.S.; Bogo, D.; Perdomo, R.T.; Antunes, A.M.; Marques, M.M.; de FC Matos, M. Synthesis and evaluation of diaryl sulfides and diaryl selenide compounds for antitubulin and cytotoxic activity. Bioorganic Med. Chem. Lett. 2013, 23, 4669–4673. [Google Scholar] [CrossRef] [Green Version]
  40. Soussi, M.A.; Provot, O.; Bernadat, G.; Bignon, J.; Wdzieczak-Bakala, J.; Desravines, D.; Dubois, J.; Brion, J.-D.; Messaoudi, S.; Alami, M. Discovery of azaisoerianin derivatives as potential antitumors agents. Eur. J. Med. Chem. 2014, 78, 178–189. [Google Scholar] [CrossRef]
  41. Herdman, C.A.; Strecker, T.E.; Tanpure, R.P.; Chen, Z.; Winters, A.; Gerberich, J.; Liu, L.; Hamel, E.; Mason, R.P.; Chaplin, D.J. Synthesis and biological evaluation of benzocyclooctene-based and indene-based anticancer agents that function as inhibitors of tubulin polymerization. MedChemComm 2016, 7, 2418–2427. [Google Scholar] [CrossRef] [Green Version]
  42. Simoni, D.; Grisolia, G.; Giannini, G.; Roberti, M.; Rondanin, R.; Piccagli, L.; Baruchello, R.; Rossi, M.; Romagnoli, R.; Invidiata, F.P. Heterocyclic and phenyl double-bond-locked combretastatin analogues possessing potent apoptosis-inducing activity in HL60 and in MDR cell lines. J. Med. Chem. 2005, 48, 723–736. [Google Scholar] [CrossRef]
  43. Sharma, S.; Kumar Gupta, M.; Kumar Saxena, A.; Singh Bedi, P.M. Thiazolidinone constraint combretastatin analogs as novel antitubulin agents: Design, synthesis, biological evaluation and docking studies. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 2017, 17, 230–240. [Google Scholar] [CrossRef]
  44. Guan, Q.; Zuo, D.; Jiang, N.; Qi, H.; Zhai, Y.; Bai, Z.; Feng, D.; Yang, L.; Jiang, M.; Bao, K. Microwave-assisted synthesis and biological evaluation of 3,4-diaryl maleic anhydride/N-substituted maleimide derivatives as combretastatin A-4 analogues. Bioorganic Med. Chem. Lett. 2015, 25, 631–634. [Google Scholar] [CrossRef] [PubMed]
  45. Barreca, M.; Spanò, V.; Raimondi, M.V.; Tarantelli, C.; Spriano, F.; Bertoni, F.; Barraja, P.; Montalbano, A. Recurrence of the oxazole motif in tubulin colchicine site inhibitors with anti-tumor activity. Eur. J. Med. Chem. Rep. 2021, 1, 100004. [Google Scholar] [CrossRef]
  46. Singh, H.; Singh, J.V.; Gupta, M.K.; Saxena, A.K.; Sharma, S.; Nepali, K.; Bedi, P.M.S. Triazole tethered isatin-coumarin based molecular hybrids as novel antitubulin agents: Design, synthesis, biological investigation and docking studies. Bioorganic Med. Chem. Lett. 2017, 27, 3974–3979. [Google Scholar] [CrossRef]
  47. Blanch, N.M.; Chabot, G.G.; Quentin, L.; Scherman, D.; Bourg, S.; Dauzonne, D. In vitro and in vivo biological evaluation of new 4,5-disubstituted 1,2,3-triazoles as cis-constrained analogs of combretastatin A4. Eur. J. Med. Chem. 2012, 54, 22–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Wang, L.; Woods, K.W.; Li, Q.; Barr, K.J.; McCroskey, R.W.; Hannick, S.M.; Gherke, L.; Credo, R.B.; Hui, Y.-H.; Marsh, K. Potent, orally active heterocycle-based combretastatin A-4 analogues: Synthesis, structure− activity relationship, pharmacokinetics, and in vivo antitumor activity evaluation. J. Med. Chem. 2002, 45, 1697–1711. [Google Scholar] [CrossRef] [PubMed]
  49. Romagnoli, R.; Baraldi, P.G.; Salvador, M.K.; Camacho, M.E.; Preti, D.; Tabrizi, M.A.; Bassetto, M.; Brancale, A.; Hamel, E.; Bortolozzi, R. Synthesis and biological evaluation of 2-substituted-4-(3′,4′,5′-trimethoxyphenyl)-5-aryl thiazoles as anticancer agents. Bioorganic Med. Chem. 2012, 20, 7083–7094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. O’Boyle, N.M.; Pollock, J.K.; Carr, M.; Knox, A.J.; Nathwani, S.M.; Wang, S.; Caboni, L.; Zisterer, D.M.; Meegan, M.J. β-Lactam estrogen receptor antagonists and a dual-targeting estrogen receptor/tubulin ligand. J. Med. Chem. 2014, 57, 9370–9382. [Google Scholar] [CrossRef] [PubMed]
  51. Greene, T.F.; Wang, S.; Greene, L.M.; Nathwani, S.M.; Pollock, J.K.; Malebari, A.M.; McCabe, T.; Twamley, B.; O’Boyle, N.M.; Zisterer, D.M. Synthesis and biochemical evaluation of 3-phenoxy-1,4-diarylazetidin-2-ones as tubulin-targeting antitumor agents. J. Med. Chem. 2016, 59, 90–113. [Google Scholar] [CrossRef]
  52. Malebari, A.M.; Duffy Morales, G.; Twamley, B.; Fayne, D.; Khan, M.F.; McLoughlin, E.C.; O’Boyle, N.M.; Zisterer, D.M.; Meegan, M.J. Synthesis, Characterisation and Mechanism of Action of Anticancer 3-Fluoroazetidin-2-ones. Pharmaceuticals 2022, 15, 1044. [Google Scholar] [CrossRef]
  53. Peng, Y.; Shi, Z.; Liang, Y.; Ding, K.; Wang, Y. Targeting the tumor microenvironment by an enzyme-responsive prodrug of tubulin destabilizer for triple-negative breast cancer therapy with high safety. Eur. J. Med. Chem. 2022, 236, 114344. [Google Scholar] [CrossRef]
  54. O’Boyle, N.M.; Carr, M.; Greene, L.M.; Bergin, O.; Nathwani, S.M.; McCabe, T.; Lloyd, D.G.; Zisterer, D.M.; Meegan, M.J. Synthesis and evaluation of azetidinone analogues of combretastatin A-4 as tubulin targeting agents. J. Med. Chem. 2010, 53, 8569–8584. [Google Scholar] [CrossRef] [PubMed]
  55. Zhou, P.; Liu, Y.; Zhou, L.; Zhu, K.; Feng, K.; Zhang, H.; Liang, Y.; Jiang, H.; Luo, C.; Liu, M. Potent antitumor activities and structure basis of the chiral β-lactam bridged analogue of combretastatin A-4 binding to tubulin. J. Med. Chem. 2016, 59, 10329–10334. [Google Scholar] [CrossRef] [PubMed]
  56. Yan, J.; Pang, Y.; Sheng, J.; Wang, Y.; Chen, J.; Hu, J.; Huang, L.; Li, X. A novel synthetic compound exerts effective anti-tumour activity in vivo via the inhibition of tubulin polymerisation in A549 cells. Biochem. Pharmacol. 2015, 97, 51–61. [Google Scholar] [CrossRef]
  57. Rasolofonjatovo, E.; Provot, O.; Hamze, A.; Rodrigo, J.; Bignon, J.; Wdzieczak-Bakala, J.; Lenoir, C.; Desravines, D.; Dubois, J.; Brion, J.-D. Design, synthesis and anticancer properties of 5-arylbenzoxepins as conformationally restricted isocombretastatin A-4 analogs. Eur. J. Med. Chem. 2013, 62, 28–39. [Google Scholar] [CrossRef] [PubMed]
  58. LeBlanc, R.; Dickson, J.; Brown, T.; Stewart, M.; Pati, H.N.; VanDerveer, D.; Arman, H.; Harris, J.; Pennington, W.; Holt, H.L., Jr. Synthesis and cytotoxicity of epoxide and pyrazole analogs of the combretastatins. Bioorganic Med. Chem. 2005, 13, 6025–6034. [Google Scholar] [CrossRef]
  59. Zaninetti, R.; Cortese, S.V.; Aprile, S.; Massarotti, A.; Canonico, P.L.; Sorba, G.; Grosa, G.; Genazzani, A.A.; Pirali, T. A Concise Synthesis of Pyrazole Analogues of Combretastatin A1 as Potent Anti-Tubulin Agents. ChemMedChem 2013, 8, 633–643. [Google Scholar] [CrossRef]
  60. Hawash, M.; Jaradat, N.; Bawwab, N.; Salem, K.; Arafat, H.; Hajyousef, Y.; Shtayeh, T.; Sobuh, S. Design, synthesis, and biological evaluation of phenyl-isoxazole-carboxamide derivatives as anticancer agents. Heterocycl. Commun. 2021, 27, 133–141. [Google Scholar] [CrossRef]
  61. Leslie, B.J.; Holaday, C.R.; Nguyen, T.; Hergenrother, P.J. Phenylcinnamides as novel antimitotic agents. J. Med. Chem. 2010, 53, 3964–3972. [Google Scholar] [CrossRef]
  62. Kamal, A.; Shaik, A.B.; Rao, B.B.; Khan, I.; Kumar, G.B.; Jain, N. Design and synthesis of pyrazole/isoxazole linked arylcinnamides as tubulin polymerization inhibitors and potential antiproliferative agents. Org. Biomol. Chem. 2015, 13, 10162–10178. [Google Scholar] [CrossRef]
  63. Ma, J.; Bao, G.; Wang, L.; Li, W.; Xu, B.; Du, B.; Lv, J.; Zhai, X.; Gong, P. Design, synthesis, biological evaluation and preliminary mechanism study of novel benzothiazole derivatives bearing indole-based moiety as potent antitumor agents. Eur. J. Med. Chem. 2015, 96, 173–186. [Google Scholar] [CrossRef]
  64. Liou, J.-P.; Wu, Z.-Y.; Kuo, C.-C.; Chang, C.-Y.; Lu, P.-Y.; Chen, C.-M.; Hsieh, H.-P.; Chang, J.-Y. Discovery of 4-amino and 4-hydroxy-1-aroylindoles as potent tubulin polymerization inhibitors. J. Med. Chem. 2008, 51, 4351–4355. [Google Scholar] [CrossRef] [PubMed]
  65. Abdellatif, K.R.; Lamie, P.F.; Omar, H.A. 3-methyl-2-phenyl-1-substituted-indole derivatives as indomethacin analogs: Design, synthesis and biological evaluation as potential anti-inflammatory and analgesic agents. J. Enzym. Inhib. Med. Chem. 2016, 31, 318–324. [Google Scholar] [CrossRef]
  66. Sharma, V.; Kumar, P.; Pathaka, D. Biological Importance of the Indole Nucleus in Recent Years: A Comprehensive Review. J. Heterocycl. Chem. 2010, 47, 491–502. [Google Scholar] [CrossRef]
  67. Zhang, M.Z.; Chen, Q.; Yang, G.F. A review on recent developments of indole-containing antiviral agents. Eur. J. Med. Chem. 2015, 89, 421–441. [Google Scholar] [CrossRef] [PubMed]
  68. Cihan-Ustundag, G.; Gursoy, E.; Naesens, L.; Ulusoy-Guzeldemirci, N.; Capan, G. Synthesis and antiviral properties of novel indole-based thiosemicarbazides and 4-thiazolidinones. Bioorganic Med. Chem. J. 2016, 24, 240–246. [Google Scholar] [CrossRef]
  69. De Martino, G.; La Regina, G.; Coluccia, A.; Edler, M.C.; Barbera, M.C.; Brancale, A.; Wilcox, E.; Hamel, E.; Artico, M.; Silvestri, R. Arylthioindoles, Potent Inhibitors of Tubulin Polymerization. J. Med. Chem. 2004, 47, 6120–6123. [Google Scholar] [CrossRef]
  70. Arthuis, M.; Pontikis, R.; Chabot, G.G.; Quentin, L.; Scherman, D.; Florent, J.C. Domino approach to 2-aroyltrimethoxyindoles as novel heterocyclic combretastatin A4 analogues. Eur. J. Med. Chem. 2011, 46, 95–100. [Google Scholar] [CrossRef]
  71. La Regina, G.; Bai, R.; Rensen, W.; Coluccia, A.; Piscitelli, F.; Gatti, V.; Bolognesi, A.; Lavecchia, A.; Granata, I.; Porta, A.; et al. Design and Synthesis of 2-Heterocyclyl-3-arylthio-1H-indoles as Potent Tubulin Polymerization and Cell Growth Inhibitors with Improved Metabolic Stability. J. Med. Chem. 2011, 54, 8394–8406. [Google Scholar] [CrossRef] [Green Version]
  72. La Regina, G.; Bai, R.; Rensen, W.M.; DíCesare, E.; Coluccia, A.; Piscitelli, F.; Famiglini, V.; Reggio, A.; Nalli, M.; Pelliccia, S.; et al. Toward highly potent cancer agents by modulating the C-2 group of the arylthioindole class of tubulin polymerization inhibitors. J. Med. Chem. 2013, 56, 123–149. [Google Scholar] [CrossRef]
  73. La Regina, G.; Edler, M.; Brancale, A.; Kandil, S.; Coluccia, A.; Piscitelli, F.; Hamel, E.; Martino, G.D.; Matesanz, R.; Dı´az, J.F.; et al. New Arythioindoles Inhibitors of Tubulin Polymerization. 3.Biological Evaluation, SAR and Molecular Modeling Studies. J. Med. Chem. 2007, 50, 749–754. [Google Scholar] [CrossRef]
  74. La Regina, G.; Sarkar, T.; Bai, R.; Edler, M.C.; Saletti, R.; Coluccia, A.; Piscitelli, F.; Minelli, L.; Gatti, V.; Mazzoccoli, C.; et al. New Arylthioindoles and Related Bioisosteres at the Sulfur Bridging Group. 4. Synthesis, Tubulin Polymerization, Cell Growth Inhibition, and Molecular Modeling Studies. J. Med. Chem. 2009, 52, 7512–7527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Tang, S.; Zhou, Z.; Jiang, Z.; Zhu, W.; Qiao, D. Indole-Based Tubulin Inhibitors: Binding Modes and SARs Investigations. Molecules 2022, 27, 1587. [Google Scholar] [CrossRef] [PubMed]
  76. Baytas, S.N. Recent Advances in Combretastatin A-4 Inspired Inhibitors of Tubulin Polymerization: An Update. Curr. Med. Chem. 2022, 29, 3557–3585. [Google Scholar] [CrossRef] [PubMed]
  77. De Martino, G.; Edler, M.C.; La Regina, G.; Coluccia, A.; Barbera, M.C.; Barrow, D.; Nicholson, R.I.; Chiosis, G.; Brancale, A.; Hamel, E. New arylthioindoles: Potent Inhibitors of tubulin polymerization. 2. Structure− activity relationships and molecular modeling studies. J. Med. Chem. 2006, 49, 947–954. [Google Scholar] [CrossRef]
  78. Alvarez, R.; Puebla, P.; Diaz, J.F.; Bento, A.C.; Garcia-Navas, R.; de la Iglesia-Vicente, J.; Mollinedo, F.; Andreu, J.M.; Medarde, M.; Pelaez, R. Endowing indole-based tubulin inhibitors with an anchor for derivatization: Highly potent 3-substituted indolephenstatins and indoleisocombretastatins. J. Med. Chem. 2013, 56, 2813–2827. [Google Scholar] [CrossRef] [Green Version]
  79. Yan, J.; Chen, J.; Zhang, S.; Hu, J.; Huang, L.; Li, X. Synthesis, evaluation, and mechanism study of novel indole-chalcone derivatives exerting effective antitumor activity through microtubule destabilization in vitro and in vivo. J. Med. Chem. 2016, 59, 5264–5283. [Google Scholar] [CrossRef]
  80. Ebenezer, O.; Shapi, M.; Tuszynski, J.A. A Review of the Recent Developments of Molecular Hybrids Targeting Tubulin Polymerization. Int. J. Mol. Sci. 2022, 23, 4001. [Google Scholar] [CrossRef]
  81. Das Mukherjee, D.; Kumar, N.M.; Tantak, M.P.; Das, A.; Ganguli, A.; Datta, S.; Kumar, D.; Chakrabarti, G. Development of Novel Bis(indolyl)-hydrazide-Hydrazone Derivatives as Potent Microtubule-Targeting Cytotoxic Agents against A549 Lung Cancer Cells. Biochemistry 2016, 55, 3020–3035. [Google Scholar] [CrossRef]
  82. Chen, H.; Deng, S.; Albadari, N.; Yun, M.-K.; Zhang, S.; Li, Y.; Ma, D.; Parke, D.N.; Yang, L.; Seagroves, T.N. Design, Synthesis, and Biological Evaluation of Stable Colchicine-Binding Site Tubulin Inhibitors 6-Aryl-2-benzoyl-pyridines as Potential Anticancer Agents. J. Med. Chem. 2021, 64, 12049–12074. [Google Scholar] [CrossRef]
  83. Mahboobi, S.; Pongratz, H.; Hufsky, H.; Hockemeyer, J.; Frieser, M.; Lyssenko, A.; Paper, D.H.; Bürgermeister, J.; Böhmer, F.-D.; Fiebig, H.-H. Synthetic 2-aroylindole derivatives as a new class of potent tubulin-inhibitory, antimitotic agents. J. Med. Chem. 2001, 44, 4535–4553. [Google Scholar] [CrossRef]
  84. Kuo, C.-C.; Hsieh, H.-P.; Pan, W.-Y.; Chen, C.-P.; Liou, J.-P.; Lee, S.-J.; Chang, Y.-L.; Chen, L.-T.; Chen, C.-T.; Chang, J.-Y. BPR0L075, a novel synthetic indole compound with antimitotic activity in human cancer cells, exerts effective antitumoral activity in vivo. Cancer Res. 2004, 64, 4621–4628. [Google Scholar] [CrossRef] [Green Version]
  85. Nien, C.-Y.; Chen, Y.-C.; Kuo, C.-C.; Hsieh, H.-P.; Chang, C.-Y.; Wu, J.-S.; Wu, S.-Y.; Liou, J.-P.; Chang, J.-Y. 5-Amino-2-aroylquinolines as highly potent tubulin polymerization inhibitors. J. Med. Chem. 2010, 53, 2309–2313. [Google Scholar] [CrossRef] [PubMed]
  86. Sharma, S.; Gupta, M.K.; Saxena, A.K.; Bedi, P.M.S. Triazole linked mono carbonyl curcumin-isatin bifunctional hybrids as novel anti tubulin agents: Design, synthesis, biological evaluation and molecular modeling studies. Bioorganic Med. Chem. 2015, 23, 7165–7180. [Google Scholar] [CrossRef] [PubMed]
  87. Haider, K.; Shafeeque, M.; Yahya, S.; Yar, M.S. A comprehensive review on pyrazoline based heterocyclic hybrids as potent anticancer agents. Eur. J. Med. Chem. Rep. 2022, 5, 100042. [Google Scholar] [CrossRef]
  88. Dutta, K.; Majumdar, A.G.; Kushwah, N.; Wadawale, A.P.; Patro, B.S.; Ghosh, S.K. Synthesis of novel indole-oxadiazole molecular hybrids by a regioselective C-3 sulfenylation of indole with 1,3,4-oxadiazole-2-thiols using iodine-dimethyl sulfoxide and their anticancer properties. J. Heterocycl. Chem. 2022, 59, 2165–2176. [Google Scholar] [CrossRef]
  89. Kamal, A.; Shaik, A.B.; Jain, N.; Kishor, C.; Nagabhushana, A.; Supriya, B.; Bharath Kumar, G.; Chourasiya, S.S.; Suresh, Y.; Mishra, R.K.; et al. Design and synthesis of pyrazole-oxindole conjugates targeting tubulin polymerization as new anticancer agents. Eur. J. Med. Chem. 2015, 92, 501–513. [Google Scholar] [CrossRef]
  90. Kamal, A.; Reddy, V.S.; Shaik, A.B.; Kumar, G.B.; Vishnuvardhan, M.V.; Polepalli, S.; Jain, N. Synthesis of (Z)-(arylamino)-pyrazolyl/isoxazolyl-2-propenones as tubulin targeting anticancer agents and apoptotic inducers. Org. Biomol. Chem. 2015, 13, 3416–3431. [Google Scholar] [CrossRef]
  91. Mowery, P.; Filkorn, M.M.; Hurysz, B.; Kwansare, D.O.; Lafferty, M.M.; McFadden, M.A.; Neerukonda, N.D.; Patel, R.R.; Pierce, K.; Sockett, K.A. Discovery of an indole-substituted furanone with tubulin polymerization inhibition activity. Bioorganic Med. Chem. Lett. 2021, 41, 127991. [Google Scholar] [CrossRef]
  92. Saruengkhanphasit, R.; Butkinaree, C.; Ornnork, N.; Lirdprapamongkol, K.; Niwetmarin, W.; Svasti, J.; Ruchirawat, S.; Eurtivong, C. Identification of new 3-phenyl-1H-indole-2-carbohydrazide derivatives and their structure–activity relationships as potent tubulin inhibitors and anticancer agents: A combined in silico, in vitro and synthetic study. Bioorganic Chem. 2021, 110, 104795. [Google Scholar] [CrossRef]
  93. Hawash, M.; Kahraman, D.C.; Cetin-Atalay, R.; Baytas, S.N. Induction of Apoptosis in Hepatocellular Carcinoma Cell Lines by Novel Indolylacrylamide Derivatives: Synthesis and Biological Evaluation. Chem. Biodivers. 2021, 18, e2001037. [Google Scholar] [CrossRef]
  94. Hawash, M.; Kahraman, D.C.; Olgac, A.; Ergun, S.G.; Hamel, E.; Cetin-Atalay, R.; Baytas, S.N. Design and Synthesis of Novel Substituted Indole-acrylamide Derivatives and Evaluation of Their Anti-Cancer Activity as Potential Tubulin-Targeting Agents. J. Mol. Struct. 2022, 1254, 132345. [Google Scholar] [CrossRef]
  95. Diao, P.-C.; Jian, X.-E.; Chen, P.; Huang, C.; Yin, J.; Huang, J.C.; Li, J.-S.; Zhao, P.-L. Design, synthesis and biological evaluation of novel indole-based oxalamide and aminoacetamide derivatives as tubulin polymerization inhibitors. Bioorganic Med. Chem. Lett. 2020, 30, 126816. [Google Scholar] [CrossRef] [PubMed]
  96. Romagnoli, R.; Baraldi, P.G.; Lopez-Cara, C.; Preti, D.; Aghazadeh Tabrizi, M.; Balzarini, J.; Bassetto, M.; Brancale, A.; Fu, X.-H.; Gao, Y. Concise synthesis and biological evaluation of 2-aroyl-5-amino benzo [b] thiophene derivatives as a novel class of potent antimitotic agents. J. Med. Chem. 2013, 56, 9296–9309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Hawash, M.M.; Kahraman, D.C.; Eren, F.; Cetin Atalay, R.; Baytas, S.N. Synthesis and biological evaluation of novel pyrazolic chalcone derivatives as novel hepatocellular carcinoma therapeutics. Eur. J. Med. Chem. 2017, 129, 12–26. [Google Scholar] [CrossRef] [PubMed]
  98. Kuchana, V.; Kashetti, V.; Tangeda, S.J.; Manga, V. Design, synthesis and molecular docking study of thiophenyl hydrazone derivatives as tubulin polymerization inhibitors. Synth. Commun. 2022, 52, 2029–2047. [Google Scholar] [CrossRef]
  99. Romagnoli, R.; Baraldi, P.G.; Kimatrai Salvador, M.; Preti, D.; Aghazadeh Tabrizi, M.; Bassetto, M.; Brancale, A.; Hamel, E.; Castagliuolo, I.; Bortolozzi, R. Synthesis and biological evaluation of 2-(alkoxycarbonyl)-3-anilinobenzo [b] thiophenes and thieno [2,3-b] pyridines as new potent anticancer agents. J. Med. Chem. 2013, 56, 2606–2618. [Google Scholar] [CrossRef] [Green Version]
  100. Penthala, N.R.; Madhukuri, L.; Thakkar, S.; Madadi, N.R.; Lamture, G.; Eoff, R.L.; Crooks, P.A. Synthesis and anti-cancer screening of novel heterocyclic-(2H)-1,2,3-triazoles as potential anti-cancer agents. MedChemComm 2015, 6, 1535–1543. [Google Scholar] [CrossRef] [Green Version]
  101. Kamal, S.; Derbala, H.A.; Alterary, S.S.; Ben Bacha, A.; Alonazi, M.; El-Ashrey, M.K.; Eid El-Sayed, N.N. Synthesis, Biological, and Molecular Docking Studies on 4,5,6,7-Tetrahydrobenzo [b] thiophene Derivatives and Their Nanoparticles Targeting Colorectal Cancer. ACS Omega 2021, 6, 28992–29008. [Google Scholar] [CrossRef]
  102. Romagnoli, R.; Preti, D.; Hamel, E.; Bortolozzi, R.; Viola, G.; Brancale, A.; Ferla, S.; Morciano, G.; Pinton, P. Concise synthesis and biological evaluation of 2-Aryl-3-Anilinobenzo [b] thiophene derivatives as potent apoptosis-inducing agents. Bioorganic Chem. 2021, 112, 104919. [Google Scholar] [CrossRef]
  103. Romagnoli, R.; Prencipe, F.; Oliva, P.; Cacciari, B.; Balzarini, J.; Liekens, S.; Hamel, E.; Brancale, A.; Ferla, S.; Manfredini, S. Synthesis and Biological Evaluation of New Antitubulin Agents Containing 2-(3′,4′,5′-trimethoxyanilino)-3,6-disubstituted-4,5,6,7-tetrahydrothieno [2,3-c] pyridine Scaffold. Molecules 2020, 25, 1690. [Google Scholar] [CrossRef]
  104. Alaylar, B.; Aygün, B.; Turhan, K.; Karadayi, G.; Şakar, E.; Singh, V.; Sayyed, M.; Pelit, E.; Karabulut, A.; Güllüce, M. Characterization of gamma-ray and neutron radiation absorption properties of synthesized quinoline derivatives and their genotoxic potential. Radiat. Phys. Chem. 2021, 184, 109471. [Google Scholar] [CrossRef]
  105. Wittmann, C.; Bacher, F.; Enyedy, E.A.; Dömötör, O.; Spengler, G.; Madejski, C.; Reynisson, J.; Arion, V.B. Highly Antiproliferative Latonduine and Indolo [2,3-c] quinoline Derivatives: Complex Formation with Copper (II) Markedly Changes the Kinase Inhibitory Profile. J. Med. Chem. 2022, 65, 2238–2261. [Google Scholar] [CrossRef] [PubMed]
  106. Mathada, B.S. The Versatile Quinoline and Its Derivatives as anti-Cancer Agents: An Overview. Polycycl. Aromat. Compd. 2022, 1–13. [Google Scholar] [CrossRef]
  107. Lai, M.-J.; Chang, J.-Y.; Lee, H.-Y.; Kuo, C.-C.; Lin, M.-H.; Hsieh, H.-P.; Chang, C.-Y.; Wu, J.-S.; Wu, S.-Y.; Shey, K.-S. Synthesis and biological evaluation of 1-(4′-Indolyl and 6′-Quinolinyl) indoles as a new class of potent anticancer agents. Eur. J. Med. Chem. 2011, 46, 3623–3629. [Google Scholar] [CrossRef] [PubMed]
  108. Li, W.; Shuai, W.; Sun, H.; Xu, F.; Bi, Y.; Xu, J.; Ma, C.; Yao, H.; Zhu, Z.; Xu, S. Design, synthesis and biological evaluation of quinoline-indole derivatives as anti-tubulin agents targeting the colchicine binding site. Eur. J. Med. Chem. 2019, 163, 428–442. [Google Scholar] [CrossRef] [PubMed]
  109. Alswah, M.; Bayoumi, A.H.; Elgamal, K.; Elmorsy, A.; Ihmaid, S.; Ahmed, H.E. Design, synthesis and cytotoxic evaluation of novel chalcone derivatives bearing triazolo [4,3-a]-quinoxaline moieties as potent anticancer agents with dual EGFR kinase and tubulin polymerization inhibitory effects. Molecules 2018, 23, 48. [Google Scholar] [CrossRef] [Green Version]
  110. Mirzaei, S.; Ghodsi, R.; Hadizadeh, F.; Sahebkar, A. 3D-QSAR-Based Pharmacophore Modeling, Virtual Screening, and Molecular Docking Studies for Identification of Tubulin Inhibitors with Potential Anticancer Activity. BioMed Res. Int. 2021, 2021, 6480804. [Google Scholar] [CrossRef]
  111. Ren, Y.; Ruan, Y.; Cheng, B.; Li, L.; Liu, J.; Fang, Y.; Chen, J. Design, synthesis and biological evaluation of novel acridine and quinoline derivatives as tubulin polymerization inhibitors with anticancer activities. Bioorganic Med. Chem. 2021, 46, 116376. [Google Scholar] [CrossRef]
  112. Al-Matarneh, M.C.; Amărandi, R.-M.; Mangalagiu, I.I.; Danac, R. Synthesis and biological screening of new cyano-substituted pyrrole fused (iso) quinoline derivatives. Molecules 2021, 26, 2066. [Google Scholar] [CrossRef]
  113. Ibrahim, T.S.; Hawwas, M.M.; Malebari, A.M.; Taher, E.S.; Omar, A.M.; Neamatallah, T.; Abdel-Samii, Z.K.; Safo, M.K.; Elshaier, Y.A. Discovery of novel quinoline-based analogues of combretastatin A-4 as tubulin polymerisation inhibitors with apoptosis inducing activity and potent anticancer effect. J. Enzym. Inhib. Med. Chem. 2021, 36, 802–818. [Google Scholar] [CrossRef]
  114. Hagras, M.; El Deeb, M.A.; Elzahabi, H.S.; Elkaeed, E.B.; Mehany, A.B.; Eissa, I.H. Discovery of new quinolines as potent colchicine binding site inhibitors: Design, synthesis, docking studies, and anti-proliferative evaluation. J. Enzym. Inhib. Med. Chem. 2021, 36, 640–658. [Google Scholar] [CrossRef] [PubMed]
  115. Ouyang, Y.; Li, J.; Chen, X.; Fu, X.; Sun, S.; Wu, Q. Chalcone Derivatives: Role in Anticancer Therapy. Biomolecules 2021, 11, 894. [Google Scholar] [CrossRef] [PubMed]
  116. Ngameni, B.; Cedric, K.; Mbaveng, A.T.; Erdoğan, M.; Simo, I.; Kuete, V.; Daştan, A. Design, synthesis, characterization, and anticancer activity of a novel series of O-substituted chalcone derivatives. Bioorganic Med. Chem. Lett. 2021, 35, 127827. [Google Scholar] [CrossRef] [PubMed]
  117. Liu, W.; He, M.; Li, Y.; Peng, Z.; Wang, G. A review on synthetic chalcone derivatives as tubulin polymerisation inhibitors. J. Enzym. Inhib. Med. Chem. 2022, 37, 9–38. [Google Scholar] [CrossRef]
  118. Sharma, S.; Kaur, C.; Budhiraja, A.; Nepali, K.; Gupta, M.K.; Saxena, A.; Bedi, P. Chalcone based azacarboline analogues as novel antitubulin agents: Design, synthesis, biological evaluation and molecular modelling studies. Eur. J. Med. Chem. 2014, 85, 648–660. [Google Scholar] [CrossRef]
  119. Oskuei, S.R.; Mirzaei, S.; Jafari-Nik, M.R.; Hadizadeh, F.; Eisvand, F.; Mosaffa, F.; Ghodsi, R. Design, synthesis and biological evaluation of novel imidazole-chalcone derivatives as potential anticancer agents and tubulin polymerization inhibitors. Bioorganic Chem. 2021, 112, 104904. [Google Scholar] [CrossRef]
  120. Wang, G.; Liu, W.; Gong, Z.; Huang, Y.; Li, Y.; Peng, Z. Design, synthesis, biological evaluation and molecular docking studies of new chalcone derivatives containing diaryl ether moiety as potential anticancer agents and tubulin polymerization inhibitors. Bioorganic Chem. 2020, 95, 103565. [Google Scholar] [CrossRef]
  121. Wang, G.; Liu, W.; Gong, Z.; Huang, Y.; Li, Y.; Peng, Z. Synthesis, biological evaluation, and molecular modelling of new naphthalene-chalcone derivatives as potential anticancer agents on MCF-7 breast cancer cells by targeting tubulin colchicine binding site. J. Enzym. Inhib. Med. Chem. 2020, 35, 139–144. [Google Scholar] [CrossRef]
  122. Wang, G.; Liu, W.; Fan, M.; He, M.; Li, Y.; Peng, Z. Design, synthesis and biological evaluation of novel thiazole-naphthalene derivatives as potential anticancer agents and tubulin polymerisation inhibitors. J. Enzym. Inhib. Med. Chem. 2021, 36, 1694–1702. [Google Scholar] [CrossRef]
  123. Sun, Y.-X.; Song, J.; Kong, L.-J.; Sha, B.-B.; Tian, X.-Y.; Liu, X.-J.; Hu, T.; Chen, P.; Zhang, S.-Y. Design, synthesis and evaluation of novel bis-substituted aromatic amide dithiocarbamate derivatives as colchicine site tubulin polymerization inhibitors with potent anticancer activities. Eur. J. Med. Chem. 2022, 229, 114069. [Google Scholar] [CrossRef]
  124. Song, J.; Wang, S.-H.; Song, C.-H.; Zhang, W.-X.; Zhu, J.-X.; Tian, X.-Y.; Fu, X.-J.; Xu, Y.; Jin, C.-Y.; Zhang, S.-Y. Discovery of N-benzylarylamide derivatives as novel tubulin polymerization inhibitors capable of activating the Hippo pathway. Eur. J. Med. Chem. 2022, 240, 114583. [Google Scholar] [CrossRef] [PubMed]
  125. Mohamed, H.S.; Amin, N.H.; El-Saadi, M.T.; Abdel-Rahman, H.M. Design, synthesis, biological assessment, and in-silico studies of 1,2,4-triazolo [1,5-a] pyrimidine derivatives as tubulin polymerization inhibitors. Bioorganic Chem. 2022, 121, 105687. [Google Scholar] [CrossRef] [PubMed]
  126. Pal, S.; Azad, A.; Bhatia, S.; Drabkin, H.; Costello, B.; Sarantopoulos, J.; Kanesvaran, R.; Lauer, R.; Starodub, A.; Hauke, R. A phase I/II trial of BNC105P with everolimus in metastatic renal cell carcinoma. Clin. Cancer Res. 2015, 21, 3420–3427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Hawash, M.; Kahraman, D.C.; Ergun, S.G.; Cetin-Atalay, R.; Baytas, S.N. Synthesis of novel indole-isoxazole hybrids and evaluation of their cytotoxic activities on hepatocellular carcinoma cell lines. BMC Chem. 2021, 15, 66. [Google Scholar] [CrossRef] [PubMed]
  128. Hao, S.-Y.; Qi, Z.-Y.; Wang, S.; Wang, X.-R.; Chen, S.-W. Synthesis and bioevaluation of N-(3,4,5-trimethoxyphenyl)-1H-pyrazolo [3,4-b] pyridin-3-amines as tubulin polymerization inhibitors with anti-angiogenic effects. Bioorganic Med. Chem. 2021, 31, 115985. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, R.; Huang, M.; Zhang, S.; Li, L.; Li, M.; Sun, J.; Wu, L.; Guan, Q.; Zhang, W. Design, synthesis and bioevaluation of 6-aryl-1-(3,4,5-trimethoxyphenyl)-1H-benzo [d] imidazoles as tubulin polymerization inhibitors. Eur. J. Med. Chem. 2021, 226, 113826. [Google Scholar] [CrossRef]
  130. Huo, X.-S.; Jian, X.-E.; Ou-Yang, J.; Chen, L.; Yang, F.; Lv, D.-X.; You, W.-W.; Rao, J.-J.; Zhao, P.-L. Discovery of highly potent tubulin polymerization inhibitors: Design, synthesis, and structure-activity relationships of novel 2,7-diaryl-[1,2,4] triazolo [1,5-a] pyrimidines. Eur. J. Med. Chem. 2021, 220, 113449. [Google Scholar] [CrossRef]
  131. Wang, B.; Wang, L.-R.; Liu, L.-L.; Wang, W.; Man, R.-J.; Zheng, D.-J.; Deng, Y.-S.; Yang, Y.-S.; Xu, C.; Zhu, H.-L. A novel series of benzothiazepine derivatives as tubulin polymerization inhibitors with anti-tumor potency. Bioorganic Chem. 2021, 108, 104585. [Google Scholar] [CrossRef]
  132. Ansari, M.; Shokrzadeh, M.; Karima, S.; Rajaei, S.; Fallah, M.; Ghassemi-Barghi, N.; Ghasemian, M.; Emami, S. New thiazole-2 (3H)-thiones containing 4-(3,4,5-trimethoxyphenyl) moiety as anticancer agents. Eur. J. Med. Chem. 2020, 185, 111784. [Google Scholar] [CrossRef]
  133. Škubník, J.; Pavlíčková, V.S.; Ruml, T.; Rimpelová, S. Vincristine in Combination Therapy of Cancer: Emerging Trends in Clinics. Biology 2021, 10, 849. [Google Scholar] [CrossRef]
  134. Wu, Q.; Qian, W.; Sun, X.; Jiang, S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J. Hematol. Oncol. 2022, 15, 143. [Google Scholar] [CrossRef] [PubMed]
  135. Alqahtani, F.Y.; Aleanizy, F.S.; El Tahir, E.; Alkahtani, H.M.; AlQuadeib, B.T. Paclitaxel. In Profiles of Drug Substances, Excipients and Related Methodology; Elsevier: Amsterdam, The Netherlands, 2019; Volume 44, pp. 205–238. [Google Scholar]
  136. Cortazar, P.; Justice, R.; Johnson, J.; Sridhara, R.; Keegan, P.; Pazdur, R. US Food and Drug Administration approval overview in metastatic breast cancer. J. Clin. Oncol. 2012, 30, 1705. [Google Scholar] [CrossRef] [PubMed]
  137. van Vuuren, R.J.; Visagie, M.H.; Theron, A.E.; Joubert, A.M. Antimitotic drugs in the treatment of cancer. Cancer Chemother. Pharmacol. 2015, 76, 1101–1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Burns, C.J.; Wilks, A.F.; Harte, M.F.; Sikanyika, H.; Fantino, E. Substituted Pyrazines as Tubulin Inhibitors. U.S. Patent No. 9,139,560, 22 September 2015. [Google Scholar]
  139. Kamal, A.; Prasad, B.; Nayak, V.L.; Reddy, V.S.; Reddy, N.V.S. N-((1-Benzyl-1H-1,2,3-triazol-4-yl)methyl) Arylamide Compounds as Potential Anticancer Agents and a Process for the Preparation Thereof. U.S. Patent No. 9309225, 12 April 2016. [Google Scholar]
  140. Thorn-Seshold, O.; Borowiak, M.; Trauner, D.; Hasserodt, J. Azoaryls as reversibly modulatable tubulin inhibitors. Google Patents 2017. [Google Scholar]
  141. Cao, X.; Li, B.; Chen, J.; Dang, J.; Chen, S.; Gunes, E.G.; Xu, B.; Tian, L.; Muend, S.; Raoof, M. Effect of cabazitaxel on macrophages improves CD47-targeted immunotherapy for triple-negative breast cancer. J. Immunother. Cancer 2021, 9, e002022. [Google Scholar] [CrossRef] [PubMed]
  142. Tong, J.T.; Harris, P.W.; Brimble, M.A.; Kavianinia, I. An Insight into FDA Approved Antibody-Drug Conjugates for Cancer Therapy. Molecules 2021, 26, 5847. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The structures of combretastatin A-4 (CA-4), fosbretabulin (CA-4P), Oxi4503 and Ombrabulin.
Figure 1. The structures of combretastatin A-4 (CA-4), fosbretabulin (CA-4P), Oxi4503 and Ombrabulin.
Biomolecules 12 01843 g001
Figure 2. The structures of CA-4 analogues when the linker is replaced with hetero atoms (Si, Se, and N).
Figure 2. The structures of CA-4 analogues when the linker is replaced with hetero atoms (Si, Se, and N).
Biomolecules 12 01843 g002
Figure 3. The structures of CA-4 analogues when the linker is replaced with heterocycles (pyrazole, tetrazole, and thiazole).
Figure 3. The structures of CA-4 analogues when the linker is replaced with heterocycles (pyrazole, tetrazole, and thiazole).
Biomolecules 12 01843 g003
Figure 4. The structures of CA-4 analogues when the linker is replaced with heterocycles (four and seven-membered rings).
Figure 4. The structures of CA-4 analogues when the linker is replaced with heterocycles (four and seven-membered rings).
Biomolecules 12 01843 g004
Figure 5. The structures of CA-4 analogues phenyl cinnamides and arylcinnamides.
Figure 5. The structures of CA-4 analogues phenyl cinnamides and arylcinnamides.
Biomolecules 12 01843 g005
Figure 6. The structures of arylthioindole, trimethoxyphenyl-indole, and bis-indole.
Figure 6. The structures of arylthioindole, trimethoxyphenyl-indole, and bis-indole.
Biomolecules 12 01843 g006
Figure 7. The structures of aroylindoles, trimethoxyphenyl-indole, indolyl-phenylmethanone, pyrazole-oxyindole, indole-amino-pyrazolyl, and indole-heterocyclic hybrid.
Figure 7. The structures of aroylindoles, trimethoxyphenyl-indole, indolyl-phenylmethanone, pyrazole-oxyindole, indole-amino-pyrazolyl, and indole-heterocyclic hybrid.
Biomolecules 12 01843 g007
Figure 8. The structures of thiophene analogues.
Figure 8. The structures of thiophene analogues.
Biomolecules 12 01843 g008
Figure 9. The structures of quinolone analogues.
Figure 9. The structures of quinolone analogues.
Biomolecules 12 01843 g009
Figure 10. The structures of chalcone derivatives.
Figure 10. The structures of chalcone derivatives.
Biomolecules 12 01843 g010
Figure 11. The structures of trimethoxyphenyl containing derivatives.
Figure 11. The structures of trimethoxyphenyl containing derivatives.
Biomolecules 12 01843 g011
Table 1. List of tubulin inhibitors in clinical developments or approved by FDA.
Table 1. List of tubulin inhibitors in clinical developments or approved by FDA.
Drug NameChemical StructureType of CancerMicrotubuleStatus
Paclitaxel [135]Biomolecules 12 01843 i001Metastatic adenocarcinoma of the pancreasStabilizingApproved in 1998
Ixabepilone [136]Biomolecules 12 01843 i002Metastatic or locally advanced breast cancerMicrotubule-stabilizingApproved in 2007
Eribulin [137]Biomolecules 12 01843 i003Recurrent metastatic breast cancerMicrotubule-destabilizingApproved in 2010
BNC105P [19]Biomolecules 12 01843 i004LeukemiaInhibit polymerizationPhase I clinical trials
Plinabulin [138]Biomolecules 12 01843 i005Lung CancerInhibit polymerizationPhase I clinical trials
Tesetaxel [139]Biomolecules 12 01843 i006Breast CancerStabilizingPhase III clinical trials
KX2-361 [140]Biomolecules 12 01843 i007Solid TumorInhibits polymerizationPhase I clinical trials
Fosbretabulin [18]Biomolecules 12 01843 i008Thyroid cancerMicrotubule-destabilizingApproved in 2018
Cabazitaxel [141]Biomolecules 12 01843 i009Metastatic, hormone-resistant prostate cancerStabilizingApproved in 2019
Monomethyl Auristatin E [142]Biomolecules 12 01843 i010Metastatic cervical cancerMicrotubule-disrupting agent (in conjugation with antibody)Approved in 2021
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hawash, M. Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy. Biomolecules 2022, 12, 1843. https://doi.org/10.3390/biom12121843

AMA Style

Hawash M. Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy. Biomolecules. 2022; 12(12):1843. https://doi.org/10.3390/biom12121843

Chicago/Turabian Style

Hawash, Mohammed. 2022. "Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy" Biomolecules 12, no. 12: 1843. https://doi.org/10.3390/biom12121843

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop