molecules-logo

Journal Browser

Journal Browser

Tubulin Inhibitors

A special issue of Molecules (ISSN 1420-3049). This special issue belongs to the section "Bioorganic Chemistry".

Deadline for manuscript submissions: closed (20 December 2016) | Viewed by 56406

Special Issue Editor


E-Mail Website
Guest Editor
Professor and Director of UTHSC College of Pharmacy Drug Discovery Center, Faculty Director of Shared Analytical Instrument Facility, University of Tennessee Health Science Center, 881 Madison Avenue, room 561, Memphis, TN 38163, USA
Interests: small molecule drug discovery; chemical biology; medicinal chemistry; tubulin inhibitors; colchicine binding site inhibitors; survivin inhibitors; MDM2 inhibitors
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Despite the recent advances of targeted therapies (mainly for cancer), chemotherapy will continue to be a broadly effective option and/or addition to targeted therapy in disease treatment. Targeting tubulin dynamics have been proven to be one of the most broadly effective methods in drug development. Currently, there are several clinically available tubulin inhibitors, including paclitaxel, docetaxel, Ixabepilone, cabazitaxel, vincristine, and colchicine. These classes of drugs have been proven to be highly useful clinically, with paclitaxel being the most widely prescribed anticancer drug.

However, there are also limitations in using many of these drugs, including toxicities associated with their systemic administration, susceptibilities to drug efflux pumps, and generally poor bioavailability. There is a constant need for access to novel tubulin inhibitors that can overcome these clinic limitations, or development of unconventional formulations that can reduce systemic toxicities to increase the therapeutic indexes of existing tubulin inhibitors. Significant advances have been made in tubulin inhibitors, with multiple new compounds undergoing clinical trials and the FDA approval of novel formulations (e.g., Abraxane, a nanoparticle formulation of paclitaxel). This Special Issue aims to provide a forum for the dissemination of the latest information on the development of new tubulin inhibitors, new formulations, increased understanding of their mechanisms, and clinical applications.

Prof. Dr. Wei Li
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Molecules is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • tubulin dynamics
  • tubulin inhibitors
  • multidrug resistance
  • nanoparticle formulations
  • beta-tubulin isotypes and drug resistance
  • angiogenesis
  • vascular disruption agent
  • antimitotic

Published Papers (6 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

4683 KiB  
Article
A 4-Phenoxyphenol Derivative Exerts Inhibitory Effects on Human Hepatocellular Carcinoma Cells through Regulating Autophagy and Apoptosis Accompanied by Downregulating α-Tubulin Expression
by Wen-Tsan Chang, Wangta Liu, Yi-Han Chiu, Bing-Hung Chen, Shih-Chang Chuang, Yen-Chun Chen, Yun-Tzh Hsu, Mei-Jei Lu, Shean-Jaw Chiou, Chon-Kit Chou and Chien-Chih Chiu
Molecules 2017, 22(5), 854; https://doi.org/10.3390/molecules22050854 - 21 May 2017
Cited by 18 | Viewed by 5969
Abstract
Hepatocellular carcinoma (HCC) is a leading cancer worldwide. Advanced HCCs are usually resistant to anticancer drugs, causing unsatisfactory chemotherapy outcomes. In this study, we showed that a 4-phenoxyphenol derivative, 4-[4-(4-hydroxyphenoxy)phenoxy]phenol (4-HPPP), exerts an inhibitory activity against two HCC cell lines, Huh7 and Ha22T. [...] Read more.
Hepatocellular carcinoma (HCC) is a leading cancer worldwide. Advanced HCCs are usually resistant to anticancer drugs, causing unsatisfactory chemotherapy outcomes. In this study, we showed that a 4-phenoxyphenol derivative, 4-[4-(4-hydroxyphenoxy)phenoxy]phenol (4-HPPP), exerts an inhibitory activity against two HCC cell lines, Huh7 and Ha22T. We further investigated the anti-HCC activities of 4-HPPP, including anti-proliferation and induction of apoptosis. Our results showed that higher dosage of 4-HPPP downregulates the expression of α-tubulin and causes nuclear enlargement in both the Huh-7 and Ha22T cell lines. Interestingly, the colony formation results showed a discrepancy in the inhibitory effect of 4-HPPP on HCC and rat liver epithelial Clone 9 cells, suggesting the selective cytotoxicity of 4-HPPP toward HCC cells. Furthermore, the cell proliferation and apoptosis assay results illustrated the differences between the two HCC cell lines. The results of cellular proliferation assays, including trypan blue exclusion and colony formation, revealed that 4-HPPP inhibits the growth of Huh7 cells, but exerts less cytotoxicity in Ha22T cells. Furthermore, the annexin V assay performed for detecting the apoptosis showed similar results. Western blotting results showed 4-HPPP caused the increase of pro-apoptotic factors including cleaved caspase-3, Bid and Bax in HCC cells, especially in Huh-7. Furthermore, an increase of autophagy-associated protein microtubule-associated protein-1 light chain-3B (LC3B)-II and the decrease of Beclin-1 and p62/SQSTM1 were observed following 4-HPPP treatment. Additionally, the level of γH2A histone family, member X (γH2AX), an endogenous DNA damage biomarker, was dramatically increased in Huh7 cells after 4-HPPP treatment, suggesting the involvement of DNA damage pathway in 4-HPPP-induced apoptosis. On the contrary, the western blotting results showed that treatment up-regulates pro-survival proteins, including the phosphorylation of protein kinase B (Akt) and the level of survivin on Ha22T cells, which may confer a resistance toward 4-HPPP. Notably, the blockade of extracellular signal-regulated kinases (ERK), but not Akt, enhanced the cytotoxicity of 4-HPPP against Ha22T cells, indicating the pro-survival role of ERK in 4-HPPP-induced anti-HCC effect. Our present work suggests that selective anti-HCC activity of 4-HPPP acts through induction of DNA damage. Accordingly, the combination of ERK inhibitor may significantly enhance the anti-cancer effect of 4-HPPP for those HCC cells which overexpress ERK in the future. Full article
(This article belongs to the Special Issue Tubulin Inhibitors)
Show Figures

Figure 1

2617 KiB  
Article
Activity of Eribulin in a Primary Culture of Well-Differentiated/Dedifferentiated Adipocytic Sarcoma
by Alessandro De Vita, Giacomo Miserocchi, Federica Recine, Laura Mercatali, Federica Pieri, Laura Medri, Alberto Bongiovanni, Davide Cavaliere, Chiara Liverani, Chiara Spadazzi, Dino Amadori and Toni Ibrahim
Molecules 2016, 21(12), 1662; https://doi.org/10.3390/molecules21121662 - 03 Dec 2016
Cited by 22 | Viewed by 5393
Abstract
Eribulin mesylate is a novel, non-taxane, synthetic microtubule inhibitor showing antitumor activity in a wide range of tumors including soft tissue sarcomas (STS). Eribulin has been recently approved for the treatment of metastatic liposarcoma (LPS) patients previously treated with anthracyclines. This work investigated [...] Read more.
Eribulin mesylate is a novel, non-taxane, synthetic microtubule inhibitor showing antitumor activity in a wide range of tumors including soft tissue sarcomas (STS). Eribulin has been recently approved for the treatment of metastatic liposarcoma (LPS) patients previously treated with anthracyclines. This work investigated the mechanism of action of this innovative antitubulin agent in well-differentiated/dedifferentiated LPS (ALT/DDLPS) which represents one of the most common adipocytic sarcoma histotypes. A primary culture of ALT/DDLPS from a 54-year-old patient was established. The anticancer activity of eribulin on the patient-derived primary culture was assessed by MTT and tunel assays. Eribulin efficacy was compared to other drugs approved for the treatment of STS. Cell migration and morphology were examined after exposure to eribulin to better understand the drug mechanism of action. Finally, Western blot analysis of apoptosis and migration proteins was performed. The results showed that eribulin exerts its antiproliferative effect by the arrest of cell motility and induction of apoptosis. Our results highlighted the activity of eribulin in the treatment of ALT/DDLPS patients. Full article
(This article belongs to the Special Issue Tubulin Inhibitors)
Show Figures

Figure 1

6944 KiB  
Article
Janus Compounds, 5-Chloro-N4-methyl-N4-aryl-9H-pyrimido[4,5-b]indole-2,4-diamines, Cause Both Microtubule Depolymerizing and Stabilizing Effects
by Cristina C. Rohena, April L. Risinger, Ravi Kumar Vyas Devambatla, Nicholas F. Dybdal-Hargreaves, Roma Kaul, Shruti Choudhary, Aleem Gangjee and Susan L. Mooberry
Molecules 2016, 21(12), 1661; https://doi.org/10.3390/molecules21121661 - 02 Dec 2016
Cited by 7 | Viewed by 5940
Abstract
While evaluating a large library of compounds designed to inhibit microtubule polymerization, we identified four compounds that have unique effects on microtubules. These compounds cause mixed effects reminiscent of both microtubule depolymerizers and stabilizers. Immunofluorescence evaluations showed that each compound initially caused microtubule [...] Read more.
While evaluating a large library of compounds designed to inhibit microtubule polymerization, we identified four compounds that have unique effects on microtubules. These compounds cause mixed effects reminiscent of both microtubule depolymerizers and stabilizers. Immunofluorescence evaluations showed that each compound initially caused microtubule depolymerization and, surprisingly, with higher concentrations, microtubule bundles were also observed. There were subtle differences in the propensity to cause these competing effects among the compounds with a continuum of stabilizing and destabilizing effects. Tubulin polymerization experiments confirmed the differential effects and, while each of the compounds increased the initial rate of tubulin polymerization at high concentrations, total tubulin polymer was not enhanced at equilibrium, likely because of the dueling depolymerization effects. Modeling studies predict that the compounds bind to tubulin within the colchicine site and confirm that there are differences in their potential interactions that might underlie their distinct effects on microtubules. Due to their dual properties of microtubule stabilization and destabilization, we propose the name Janus for these compounds after the two-faced Roman god. The identification of synthetically tractable, small molecules that elicit microtubule stabilizing effects is a significant finding with the potential to identify new mechanisms of microtubule stabilization. Full article
(This article belongs to the Special Issue Tubulin Inhibitors)
Show Figures

Graphical abstract

Review

Jump to: Research

5500 KiB  
Review
Tubulin Inhibitor-Based Antibody-Drug Conjugates for Cancer Therapy
by Hao Chen, Zongtao Lin, Kinsie E. Arnst, Duane D. Miller and Wei Li
Molecules 2017, 22(8), 1281; https://doi.org/10.3390/molecules22081281 - 01 Aug 2017
Cited by 138 | Viewed by 17088
Abstract
Antibody-drug conjugates (ADCs) are a class of highly potent biopharmaceutical drugs generated by conjugating cytotoxic drugs with specific monoclonal antibodies through appropriate linkers. Specific antibodies used to guide potent warheads to tumor tissues can effectively reduce undesired side effects of the cytotoxic drugs. [...] Read more.
Antibody-drug conjugates (ADCs) are a class of highly potent biopharmaceutical drugs generated by conjugating cytotoxic drugs with specific monoclonal antibodies through appropriate linkers. Specific antibodies used to guide potent warheads to tumor tissues can effectively reduce undesired side effects of the cytotoxic drugs. An in-depth understanding of antibodies, linkers, conjugation strategies, cytotoxic drugs, and their molecular targets has led to the successful development of several approved ADCs. These ADCs are powerful therapeutics for cancer treatment, enabling wider therapeutic windows, improved pharmacokinetic/pharmacodynamic properties, and enhanced efficacy. Since tubulin inhibitors are one of the most successful cytotoxic drugs in the ADC armamentarium, this review focuses on the progress in tubulin inhibitor-based ADCs, as well as lessons learned from the unsuccessful ADCs containing tubulin inhibitors. This review should be helpful to facilitate future development of new generations of tubulin inhibitor-based ADCs for cancer therapy. Full article
(This article belongs to the Special Issue Tubulin Inhibitors)
Show Figures

Figure 1

1119 KiB  
Review
Current Advances of Tubulin Inhibitors in Nanoparticle Drug Delivery and Vascular Disruption/Angiogenesis
by Souvik Banerjee, Dong-Jin Hwang, Wei Li and Duane D. Miller
Molecules 2016, 21(11), 1468; https://doi.org/10.3390/molecules21111468 - 02 Nov 2016
Cited by 44 | Viewed by 9972
Abstract
Extensive research over the last decade has resulted in a number of highly potent tubulin polymerization inhibitors acting either as microtubule stabilizing agents (MSAs) or microtubule destabilizing agents (MDAs). These inhibitors have potent cytotoxicity against a broad spectrum of human tumor cell lines. [...] Read more.
Extensive research over the last decade has resulted in a number of highly potent tubulin polymerization inhibitors acting either as microtubule stabilizing agents (MSAs) or microtubule destabilizing agents (MDAs). These inhibitors have potent cytotoxicity against a broad spectrum of human tumor cell lines. In addition to cytotoxicity, a number of these tubulin inhibitors have exhibited abilities to inhibit formation of new blood vessels as well as disrupt existing blood vessels. Tubulin inhibitors as a vascular disrupting agents (VDAs), mainly from the MDA family, induce rapid tumor vessel occlusion and massive tumor necrosis. Thus, tubulin inhibitors have become increasingly popular in the field of tumor vasculature. However, their pharmaceutical application is halted by a number of limitations including poor solubility and toxicity. Thus, recently, there has been considerable interests in the nanoparticle drug delivery of tubulin inhibitors to circumvent those limitations. This article reviews recent advances in nanoparticle based drug delivery for tubulin inhibitors as well as their tumor vasculature disruption properties. Full article
(This article belongs to the Special Issue Tubulin Inhibitors)
Show Figures

Figure 1

6545 KiB  
Review
Novel Natural Product- and Privileged Scaffold-Based Tubulin Inhibitors Targeting the Colchicine Binding Site
by Mengqi Dong, Fang Liu, Hongyu Zhou, Shumei Zhai and Bing Yan
Molecules 2016, 21(10), 1375; https://doi.org/10.3390/molecules21101375 - 15 Oct 2016
Cited by 63 | Viewed by 10869
Abstract
Tubulin inhibitors are effective anticancer agents, however, there are many limitations to the use of available tubulin inhibitors in the clinic, such as multidrug resistance, severe side-effects, and generally poor bioavailability. Thus, there is a constant need to search for novel tubulin inhibitors [...] Read more.
Tubulin inhibitors are effective anticancer agents, however, there are many limitations to the use of available tubulin inhibitors in the clinic, such as multidrug resistance, severe side-effects, and generally poor bioavailability. Thus, there is a constant need to search for novel tubulin inhibitors that can overcome these limitations. Natural product and privileged structures targeting tubulin have promoted the discovery and optimization of tubulin inhibitors. This review will focus on novel tubulin inhibitors derived from natural products and privileged structures targeting the colchicine binding site on tubulin. Full article
(This article belongs to the Special Issue Tubulin Inhibitors)
Show Figures

Figure 1

Back to TopTop