51
|
Yele V, Pindiprolu SKSS, Sana S, Ramamurty DSVNM, Madasi JRK, Vadlamani S. Synthesis and Preclinical Evaluation of Indole Triazole Conjugates as Microtubule Targeting Agents that are Effective against MCF-7 Breast Cancer Cell Lines. Anticancer Agents Med Chem 2021; 21:1047-1055. [PMID: 32981511 DOI: 10.2174/1871520620666200925102940] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 11/22/2022]
Abstract
CDATA[Background: Microtubules are considered to be an important therapeutic target for most of the anticancer drugs. These are highly dynamic structures comprising of α-tubulin and β-tubulin which are usually heterodimers and found to be involved in cell movement, intracellular trafficking, and mitosis inhibition of which might kill the tumour cells or inhibit the abnormal proliferation of cells. Most of the tubulin polymerization inhibitors, such as Vinca alkaloids, consist of Indole as the main scaffold. The literature also suggests using triazole moiety in the chemical entities, potentiating the inhibitory activity against cell proliferation. So, in our study, we used indole triazole scaffolds to synthesize the derivatives against tubulin polymerization. OBJECTIVE The main objective of this study to synthesize indole triazole conjugates by using environmentally friendly solvents (green chemistry) and click chemistry. To carry out the MTT assay and tubulin polymerization assay for the synthesized indole triazole conjugates. METHODS All the synthesized molecules were subjected to molecular docking studies using Schrodinger suite and the structural confirmation was performed by Mass, proton-NMR and carbon-NMR, documented in DMSO and CDCL3. Biological studies were performed using DU145 (prostate cancer), A-549 (lung cancer) and, MCF-7 (breast cancer), cell lines obtained from ATCC were maintained as a continuous culture. MTT assay was performed for the analogues using standard protocol. Cell cycle analysis was carried out using flow cytometry. RESULTS The Indole triazole scaffolds were synthesized using the principles of Green chemistry. The triazole formation is mainly achieved by using the Click chemistry approach. Structural elucidation of synthesized compounds was performed using Mass spectroscopy (HR-MS), Proton-Nuclear Magnetic Spectroscopy (1H-NMR) and Carbon-Nuclear Magnetic Spectroscopy (13C-NMR). The XP-docked poses and free energy binding calculations revealed that 2c and 2g molecules exhibited the highest docking affinity against the tubulin-colchicine domain (PDB:1SA0). In vitro cytotoxic assessment revealed that 2c and 2g displayed promising cytotoxicity in MTT assay (with CTC50 values 3.52μM and 2.37μM) which are in good agreement with the computational results. 2c and 2g also arrested 63 and 66% of cells in the G2/M phase, respectively, in comparison to control cells (10%) and tubulin polymerization inhibition assay revealed that 2c and 2g exhibited significant inhibition of tubulin polymerization with IC50 values of 2.31μM, and 2.62μM, respectively in comparison to Nocodazole, a positive control, resulted in an IC50 value of 2.51μM. CONCLUSION Indole triazole hybrids were synthesized using click chemistry, and docking studies were carried out using Schrodinger for the designed molecules. Process Optimization has been done for both the schemes. Twelve compounds (2a-2l) have been successfully synthesized and analytical evaluation was performed using NMR and HR-MS. In vitro evaluation was for the synthesized molecules to check tubulin polymerization inhibition for antiproliferative action. Among the synthesized compounds, 2c and 2g have potent anticancer activities by inhibiting tubulin polymerization.
Collapse
Affiliation(s)
- Vidyasrilekha Yele
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamilnadu, India
| | - Sravani Sana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - D S V N M Ramamurty
- Department of Chemistry, Government Degree College (A), Tuni, Andhra Pradesh, India
| | - Jayanthi R K Madasi
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Swapna Vadlamani
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| |
Collapse
|
52
|
Ren Y, Wang Y, Li G, Zhang Z, Ma L, Cheng B, Chen J. Discovery of Novel Benzimidazole and Indazole Analogues as Tubulin Polymerization Inhibitors with Potent Anticancer Activities. J Med Chem 2021; 64:4498-4515. [PMID: 33788562 DOI: 10.1021/acs.jmedchem.0c01837] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Novel indazole and benzimidazole analogues were designed and synthesized as tubulin inhibitors with potent antiproliferative activities. Among them, compound 12b exhibited the strongest inhibitory effects on the growth of cancer cells with an average IC50 value of 50 nM, slightly better than colchicine. 12b exhibited nearly equal potency against both, a paclitaxel-resistant cancer cell line (A2780/T, IC50 = 9.7 nM) and the corresponding parental cell line (A2780S, IC50 = 6.2 nM), thus effectively overcoming paclitaxel resistance in vitro. The crystal structure of 12b in complex with tubulin was solved to 2.45 Å resolution by X-ray crystallography, and its direct binding was confirmed to the colchicine site. Furthermore, 12b displayed significant in vivo antitumor efficacy in a melanoma tumor model with tumor growth inhibition rates of 78.70% (15 mg/kg) and 84.32% (30 mg/kg). Collectively, this work shows that 12b is a promising lead compound deserving further investigation as a potential anticancer agent.
Collapse
Affiliation(s)
- Yichang Ren
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Precision Medicine Research Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Zherong Zhang
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Lingling Ma
- Targeted Tracer Research and Development Laboratory, Precision Medicine Research Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Binbin Cheng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
53
|
Dixit H, Kumar C S, Chaudhary R, Thaker D, Gadewal N, Dasgupta D. Role of Phosphorylation and Hyperphosphorylation of Tau in Its Interaction with βα Dimeric Tubulin Studied from a Bioinformatics Perspective. Avicenna J Med Biotechnol 2021; 13:24-34. [PMID: 33680370 PMCID: PMC7903436 DOI: 10.18502/ajmb.v13i1.4579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background: Tau is a disordered Microtubule Associated Protein (MAP) which prefers to bind and stabilize microtubules. Phosphorylation of tau in particular enhances tautubulin interaction which otherwise detaches from tubulin during hyperphosphorylation. The reason behind their destabilization, detachment and the role of β subunit (from microtubule) and the projection domain (Tau) in microtubule stability remains elusive till date. Thus, a complete 3D structural investigation of tau protein is much needed to address these queries as the existing crystal structures are in fragments and quite limited. Methods: In this study, the modelled human tau protein was subjected to phosphorylation and hyperphosphorylation which were later considered for docking with micro-tubules (βα subunits-inter dimer) and vinblastine. Results: Phosphorylated tau protein interacts with both α- and β subunits. But stronger bonding was with α- compared to β subunits. Regarding β subunit, proline rich loop and projection domain actively participated in tau binding. Interestingly, hyperphosphorylation of tau increases MAP domain flexibility which ultimately results in tau detachment, the main reason behind tangle formation in Alzheimer’s disease. Conclusion: This study being the first of its kind emphasizes the role of projection domain and proline rich region of β-subunit in stabilizing the tau-tubulin interaction and also the effect of hyperphosphorylation in protein-protein and protein-drug binding.
Collapse
Affiliation(s)
- Hrushikesh Dixit
- Faculty of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India
| | - Selvaa Kumar C
- Faculty of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India
| | - Ruchi Chaudhary
- Faculty of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India
| | - Divya Thaker
- Faculty of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India
| | - Nikhil Gadewal
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Kharghar, Navi Mumbai, India
| | - Debjani Dasgupta
- Faculty of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India
| |
Collapse
|
54
|
Boichuk S, Galembikova A, Bikinieva F, Dunaev P, Aukhadieva A, Syuzov K, Zykova S, Igidov N, Ksenofontov A, Bocharov P. 2-APCAs, the Novel Microtubule Targeting Agents Active Against Distinct Cancer Cell Lines. Molecules 2021; 26:616. [PMID: 33503939 PMCID: PMC7865999 DOI: 10.3390/molecules26030616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 11/17/2022] Open
Abstract
Microtubules are known as the most attractive molecular targets for anti-cancer drugs. However, the number of serious limitations of the microtubule targeting agents (MTAs) including poor bioavailability, adverse effects (e.g., systemic and neural toxicity), and acquired resistance after initiation of MTA-based therapy remain the driving forces to develop the novel therapeutic agents effectively targeting microtubules and exhibiting potent anti-tumor activities. Here, we report the discovery of 2-amino-pyrrole-carboxamides (2-APCAs), a novel class of MTA, which effectively inhibited the growth of the broad spectrum of cancer cell lines in vitro, including various types of breast, prostate, and non-small lung cancer (NSLC), soft tissue sarcomas (STS) (e.g., leio-, rhabdomyo-, and fibrosarcomas), osteosarcomas and gastrointestinal stromal tumors (GISTs). Importantly, 2-APCAs were also effective in cancer cell lines exhibiting resistance to certain chemotherapeutic agents, including MTAs and topoisomerase II inhibitors. The anti-proliferative effect of 2-APCAs was due to their ability to interfere with the polymerization of tubulin and thereby leading to the accumulation of tumor cells in the M-phase. As an outcome of the mitotic arrest, cancer cells underwent apoptotic cell death which was evidenced by increased expression of cleaved forms of the poly-ADP-ribose polymerase (PARP) and caspase-3 and the increased numbers of Annexin V-positive cells, as well. Among the compounds exhibiting the potent anti-cancer activities against the various cancer cell lines indicated above, 2-APCA-III was found the most active. Importantly, its cytotoxic activities correlated with its highest potency to interfere with the dynamics of tubulin polymerization and inducement of cell cycle arrest in the G2/M phase. Interestingly, the cytotoxic and tubulin polymerization activities of 2-APCAs correlated with the stability of the «tubulin-2-АРСА» complexes, illustrating the "tubulin-2-APCA-III" complex as the most stable. Molecular docking showed that the binding site for 2-АРСА-III is located in α tubulin by forming a hydrogen bond with Leu23. Of note, single-cell electrophoresis (Comet assay) data illustrated the low genotoxic activities of 2-APCAs when compared to certain anti-cancer chemotherapeutic agents. Taken together, our study describes the novel MTAs with potent anti-proliferative and pro-apoptotic activities, thereby illustrating them as a scaffold for the development of successful chemotherapeutic anti-cancer agent targeting microtubules.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (A.G.); (F.B.); (P.D.); (A.A.); (K.S.)
- Сentral Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (A.G.); (F.B.); (P.D.); (A.A.); (K.S.)
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (A.G.); (F.B.); (P.D.); (A.A.); (K.S.)
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (A.G.); (F.B.); (P.D.); (A.A.); (K.S.)
| | - Aida Aukhadieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (A.G.); (F.B.); (P.D.); (A.A.); (K.S.)
| | - Kirill Syuzov
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (A.G.); (F.B.); (P.D.); (A.A.); (K.S.)
| | - Svetlana Zykova
- Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (N.I.)
| | - Nazim Igidov
- Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (N.I.)
| | - Alexander Ksenofontov
- G.A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, 153045 Ivanovo, Russia; (A.K.); (P.B.)
| | - Pavel Bocharov
- G.A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, 153045 Ivanovo, Russia; (A.K.); (P.B.)
- Institute of Solution Chemistry, Ivanovo State University of Chemistry and Technology, 153000 Ivanovo, Russia
| |
Collapse
|
55
|
Xiang W, Quadery TM, Hamel E, Luckett-Chastain LR, Ihnat MA, Mooberry SL, Gangjee A. The 3-D conformational shape of N-naphthyl-cyclopenta[d]pyrimidines affects their potency as microtubule targeting agents and their antitumor activity. Bioorg Med Chem 2021; 29:115887. [PMID: 33310545 PMCID: PMC7875120 DOI: 10.1016/j.bmc.2020.115887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 01/18/2023]
Abstract
A series of methoxy naphthyl substituted cyclopenta[d]pyrimidine compounds, 4-10, were designed and synthesized to study the influence of the 3-D conformation on microtubule depolymerizing and antiproliferative activities. NOESY studies with the N,2-dimethyl-N-(6'-methoxynaphthyl-1'-amino)-cyclopenta[d]pyrimidin-4-amine (4) showed hindered rotation of the naphthyl ring around the cyclopenta[d]pyrimidine scaffold. In contrast, NOESY studies with N,2-dimethyl-N-(5'-methoxynaphthyl-2'-amino)-cyclopenta[d]pyrimidin-4-amine (5) showed free rotation of the naphthyl ring around the cyclopenta[d]pyrimidine scaffold. The rotational flexibility and conformational dissimilarity between 4 and 5 led to a significant difference in biological activities. Compound 4 is inactive while 5 is the most potent in this series with potent microtubule depolymerizing effects and low nanomolar IC50 values in vitro against a variety of cancer cell lines. The ability of 5 to inhibit tumor growth in vivo was investigated in a U251 glioma xenograft model. The results show that 5 had better antitumor effects than the positive control temozolomide and have identified 5 as a potential preclinical candidate for further studies. The influence of conformation on the microtubule depolymerizing and antitumor activity forms the basis for the development of conformation-activity relationships for the cyclopenta[d]pyrimidine class of microtubule targeting agents.
Collapse
Affiliation(s)
- Weiguo Xiang
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Tasdique M Quadery
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, United States
| | - Lerin R Luckett-Chastain
- Department of Pharmaceutical Sciences, The University of Oklahoma College of Pharmacy, Oklahoma City, OK 73117, United States
| | - Michael A Ihnat
- Department of Pharmaceutical Sciences, The University of Oklahoma College of Pharmacy, Oklahoma City, OK 73117, United States
| | - Susan L Mooberry
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States.
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States.
| |
Collapse
|
56
|
Avila C, Angulo-Preckler C. Bioactive Compounds from Marine Heterobranchs. Mar Drugs 2020; 18:657. [PMID: 33371188 PMCID: PMC7767343 DOI: 10.3390/md18120657] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022] Open
Abstract
The natural products of heterobranch molluscs display a huge variability both in structure and in their bioactivity. Despite the considerable lack of information, it can be observed from the recent literature that this group of animals possesses an astonishing arsenal of molecules from different origins that provide the molluscs with potent chemicals that are ecologically and pharmacologically relevant. In this review, we analyze the bioactivity of more than 450 compounds from ca. 400 species of heterobranch molluscs that are useful for the snails to protect themselves in different ways and/or that may be useful to us because of their pharmacological activities. Their ecological activities include predator avoidance, toxicity, antimicrobials, antifouling, trail-following and alarm pheromones, sunscreens and UV protection, tissue regeneration, and others. The most studied ecological activity is predation avoidance, followed by toxicity. Their pharmacological activities consist of cytotoxicity and antitumoral activity; antibiotic, antiparasitic, antiviral, and anti-inflammatory activity; and activity against neurodegenerative diseases and others. The most studied pharmacological activities are cytotoxicity and anticancer activities, followed by antibiotic activity. Overall, it can be observed that heterobranch molluscs are extremely interesting in regard to the study of marine natural products in terms of both chemical ecology and biotechnology studies, providing many leads for further detailed research in these fields in the near future.
Collapse
Affiliation(s)
- Conxita Avila
- Department of Evolutionary Biology, Ecology, and Environmental Sciences, Biodiversity Research Institute (IrBIO), Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Catalonia, Spain;
| | - Carlos Angulo-Preckler
- Department of Evolutionary Biology, Ecology, and Environmental Sciences, Biodiversity Research Institute (IrBIO), Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Catalonia, Spain;
- Norwegian College of Fishery Science, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9019 Tromsø, Norway
| |
Collapse
|
57
|
High-resolution X-ray structure of three microtubule-stabilizing agents in complex with tubulin provide a rationale for drug design. Biochem Biophys Res Commun 2020; 534:330-336. [PMID: 33272565 DOI: 10.1016/j.bbrc.2020.11.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 02/05/2023]
Abstract
Microtubule is a key component of cytoskeleton and has been considered as an important target for the treatment of cancer. In particular, the tubulin taxane-site inhibitors such as taxol analogs and epothilones have achieved great success in clinical trials. However, the structural basis of many taxane-site inhibitors is still lacking in exploring their mechanism of action. We here reported crystal complex structures for three taxane-site inhibitors, Ixabepilone, Epothilone B, and Epothilone D, which were determined to 2.4 Å, 2.4 Å, and 2.85 Å, respectively. The crystal structures revealed that these taxane-site inhibitors possess similar binding modes to that of Epothilone A at the taxane site, e.g. making critical hydrogen-bonding interactions with multiple residues on the M-loop, which facilitating the tubulin polymerization. Furthermore, we summarized the binding modes of almost all taxane-site inhibitors and identified novel taxane-site ligands with simpler chemical structures through virtual screening. On this basis, new derivatives with higher binding affinity to tubulin were designed and developed, which can form additional hydrogen bond interactions with tubulin. Overall, this work determined the mechanism of action of epothilones and provided a structural basis to design reasonably novel taxane-site inhibitors with simpler structure and improved pharmacokinetic properties.
Collapse
|
58
|
Škubník J, Jurášek M, Ruml T, Rimpelová S. Mitotic Poisons in Research and Medicine. Molecules 2020; 25:E4632. [PMID: 33053667 PMCID: PMC7587177 DOI: 10.3390/molecules25204632] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the greatest challenges of the modern medicine. Although much effort has been made in the development of novel cancer therapeutics, it still remains one of the most common causes of human death in the world, mainly in low and middle-income countries. According to the World Health Organization (WHO), cancer treatment services are not available in more then 70% of low-income countries (90% of high-income countries have them available), and also approximately 70% of cancer deaths are reported in low-income countries. Various approaches on how to combat cancer diseases have since been described, targeting cell division being among them. The so-called mitotic poisons are one of the cornerstones in cancer therapies. The idea that cancer cells usually divide almost uncontrolled and far more rapidly than normal cells have led us to think about such compounds that would take advantage of this difference and target the division of such cells. Many groups of such compounds with different modes of action have been reported so far. In this review article, the main approaches on how to target cancer cell mitosis are described, involving microtubule inhibition, targeting aurora and polo-like kinases and kinesins inhibition. The main representatives of all groups of compounds are discussed and attention has also been paid to the presence and future of the clinical use of these compounds as well as their novel derivatives, reviewing the finished and ongoing clinical trials.
Collapse
Affiliation(s)
- Jan Škubník
- Department of Biochemistry and Microbiology, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic; (J.Š.); (T.R.)
| | - Michal Jurášek
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic;
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic; (J.Š.); (T.R.)
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic; (J.Š.); (T.R.)
| |
Collapse
|
59
|
Su M, Zhao C, Li D, Cao J, Ju Z, Kim EL, Jung YS, Jung JH. Viriditoxin Stabilizes Microtubule Polymers in SK-OV-3 Cells and Exhibits Antimitotic and Antimetastatic Potential. Mar Drugs 2020; 18:md18090445. [PMID: 32867174 PMCID: PMC7551567 DOI: 10.3390/md18090445] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Microtubules play a crucial role in mitosis and are attractive targets for cancer therapy. Recently, we isolated viriditoxin, a cytotoxic and antibacterial compound, from a marine fungus Paecilomyces variotii. Viriditoxin has been reported to inhibit the polymerization of bacterial FtsZ, a tubulin-like GTPase that plays an essential role in bacterial cell division. Given the close structural homology between FtsZ and tubulin, we investigated the potential antimitotic effects of viriditoxin on human cancer cells. Viriditoxin, like paclitaxel, enhanced tubulin polymerization and stabilized microtubule polymers, thereby perturbing mitosis in the SK-OV-3 cell line. However, the morphology of the stabilized microtubules was different from that induced by paclitaxel, indicating subtle differences in the mode of action of these compounds. Microtubule dynamics are also essential in cell movement, and viriditoxin repressed migration and colony formation ability of SK-OV-3 cells. Based on these results, we propose that viriditoxin interrupts microtubule dynamics, thus leading to antimitotic and antimetastatic activities.
Collapse
Affiliation(s)
- Mingzhi Su
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Changhao Zhao
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Dandan Li
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Jiafu Cao
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Zhiran Ju
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Eun La Kim
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Jee H. Jung
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
- Correspondence:
| |
Collapse
|
60
|
Borys F, Joachimiak E, Krawczyk H, Fabczak H. Intrinsic and Extrinsic Factors Affecting Microtubule Dynamics in Normal and Cancer Cells. Molecules 2020; 25:E3705. [PMID: 32823874 PMCID: PMC7464520 DOI: 10.3390/molecules25163705] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/18/2022] Open
Abstract
Microtubules (MTs), highly dynamic structures composed of α- and β-tubulin heterodimers, are involved in cell movement and intracellular traffic and are essential for cell division. Within the cell, MTs are not uniform as they can be composed of different tubulin isotypes that are post-translationally modified and interact with different microtubule-associated proteins (MAPs). These diverse intrinsic factors influence the dynamics of MTs. Extrinsic factors such as microtubule-targeting agents (MTAs) can also affect MT dynamics. MTAs can be divided into two main categories: microtubule-stabilizing agents (MSAs) and microtubule-destabilizing agents (MDAs). Thus, the MT skeleton is an important target for anticancer therapy. This review discusses factors that determine the microtubule dynamics in normal and cancer cells and describes microtubule-MTA interactions, highlighting the importance of tubulin isoform diversity and post-translational modifications in MTA responses and the consequences of such a phenomenon, including drug resistance development.
Collapse
Affiliation(s)
- Filip Borys
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| | - Hanna Krawczyk
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| |
Collapse
|
61
|
Kim J, Lee J, Lee J, Keum H, Kim Y, Kim Y, Yu B, Lee SY, Tanaka J, Jon S, Choi MC. Tubulin-Based Nanotubes as Delivery Platform for Microtubule-Targeting Agents. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002902. [PMID: 32579276 DOI: 10.1002/adma.202002902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Indexed: 06/11/2023]
Abstract
Tubulin-based nanotubes (TNTs) to deliver microtubule-targeting agents (MTAs) for clinical oncology are reported. Three MTAs, docetaxel (DTX), laulimalide (LMD), and monomethyl auristatin E (MMAE), which attach to different binding sites in a tubulin, are loaded onto TNTs and cause structural changes in them, including shape anisotropy and tubulin layering. This drug-driven carrier transformation leads to changes in the drug-loading efficiency and stability characteristics of the carrier. TNTs coloaded with DTX and LMD efficiently deliver dual drug cargoes to cellular tubulins by the endolysosomal pathway, and results in synergistic anticancer and antiangiogenic action of the drugs in vitro. In in vivo tests, TNTs loaded with a microtubule-destabilizing agent MMAE suppress the growth of tumors with much higher efficacy than free MMAE did. This work suggests a new concept of using a drug's target protein as a carrier. The findings demonstrate that the TNTs developed here can be used universally as a delivery platform for many MTAs.
Collapse
Affiliation(s)
- Jinjoo Kim
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Juncheol Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Jimin Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Hyeongseop Keum
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Yumi Kim
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Yujin Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Byeongjun Yu
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Sang Yeop Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Junichi Tanaka
- Department of Chemistry, Biology and Marine Science, University of the Ryukyus, Nishihara, Okinawa, 903-0213, Japan
| | - Sangyong Jon
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| | - Myung Chul Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Korea
| |
Collapse
|
62
|
Risinger AL, Du L. Targeting and extending the eukaryotic druggable genome with natural products: cytoskeletal targets of natural products. Nat Prod Rep 2020; 37:634-652. [PMID: 31764930 PMCID: PMC7797185 DOI: 10.1039/c9np00053d] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Covering: 2014-2019We review recent progress on natural products that target cytoskeletal components, including microtubules, actin, intermediate filaments, and septins and highlight their demonstrated and potential utility in the treatment of human disease. The anticancer efficacy of microtubule targeted agents identified from plants, microbes, and marine organisms is well documented. We highlight new microtubule targeted agents currently in clinical evaluations for the treatment of drug resistant cancers and the accumulating evidence that the anticancer efficacy of these agents is not solely due to their antimitotic effects. Indeed, the effects of microtubule targeted agents on interphase microtubules are leading to their potential for more mechanistically guided use in cancers as well as neurological disease. The discussion of these agents as more targeted drugs also prompts a reevaluation of our thinking about natural products that target other components of the cytoskeleton. For instance, actin active natural products are largely considered chemical probes and non-selective toxins. However, studies utilizing these probes have uncovered aspects of actin biology that can be more specifically targeted to potentially treat cancer, neurological disorders, and infectious disease. Compounds that target intermediate filaments and septins are understudied, but their continued discovery and mechanistic evaluations have implications for numerous therapeutic indications.
Collapse
Affiliation(s)
- April L Risinger
- The University of Texas Health Science Center at San Antonio, Department of Pharmacology, 7703 Floyd Curl Drive, San Antonio, Texas 78229, USA.
| | | |
Collapse
|
63
|
Čermák V, Dostál V, Jelínek M, Libusová L, Kovář J, Rösel D, Brábek J. Microtubule-targeting agents and their impact on cancer treatment. Eur J Cell Biol 2020; 99:151075. [PMID: 32414588 DOI: 10.1016/j.ejcb.2020.151075] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule-targeting agents (MTAs) constitute a diverse group of chemical compounds that bind to microtubules and affect their properties and function. Disruption of microtubules induces various cellular responses often leading to cell cycle arrest or cell death, the most common effect of MTAs. MTAs have found a plethora of practical applications in weed control, as fungicides and antiparasitics, and particularly in cancer treatment. Here we summarize the current knowledge of MTAs, the mechanisms of action and their role in cancer treatment. We further outline the potential use of MTAs in anti-metastatic therapy based on inhibition of cancer cell migration and invasiveness. The two main problems associated with cancer therapy by MTAs are high systemic toxicity and development of resistance. Toxic side effects of MTAs can be, at least partly, eliminated by conjugation of the drugs with various carriers. Moreover, some of the novel MTAs overcome the resistance mediated by both multidrug resistance transporters as well as overexpression of specific β-tubulin types. In anti-metastatic therapy, MTAs should be combined with other drugs to target all modes of cancer cell invasion.
Collapse
Affiliation(s)
- Vladimír Čermák
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Vojtěch Dostál
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Michael Jelínek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Libusová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Jan Kovář
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic.
| |
Collapse
|
64
|
Zúñiga-Bustos M, Vásquez PA, Jaña GA, Guzmán JL, Alderete JB, Jiménez VA. Mechanism-Based Rational Discovery and In Vitro Evaluation of Novel Microtubule Stabilizing Agents with Non-Taxol-Competitive Activity. J Chem Inf Model 2020; 60:3204-3213. [PMID: 32286822 DOI: 10.1021/acs.jcim.9b01133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Microtubules (MT) are cytoskeletal polymers of αβ-tubulin dimers that play a critical role in many cellular functions. Diverse antimitotic drugs bind to MT and disrupt their dynamics acting as MT stabilizing or destabilizing agents. The occurrence of undesired side effects and drug resistance encourages the search for novel MT binding agents with chemically diverse structures and different interaction profiles compared to known active compounds. This work reports the rational discovery of seven novel MT stabilizers using a combination of molecular modeling methods and in vitro experimental assays. Virtual screening, similarity filtering, and molecular mechanics generalized Born surface area (MM/GBSA) binding free energy refinement were employed to select seven potential candidates with high predicted affinity toward the non-taxoid site for MT stabilizers on β-tubulin. MD simulations of 150 ns on reduced MT models suggest that candidate compounds strengthen the longitudinal interactions between tubulin dimers across protofilaments, which is a primary molecular mechanism of action for known MT stabilizers. In vitro MT polymerization assays confirmed that all candidates promote MT assembly at concentrations of >50 mM and exhibit noncompetitive MT polymerization profiles when cotreating with Taxol. Preliminary HeLa cell viability assays revealed a moderate cytotoxic effect for the compounds under study at 100 μM concentration. These results support the validity of our rational discovery strategy and the use of molecular modeling methods to pursue the search and optimization of new MT targeting agents.
Collapse
Affiliation(s)
- Matías Zúñiga-Bustos
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano 4260000, Chile
| | - Pilar A Vásquez
- Laboratory of Molecular Neurobiology, Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepción 4070386, Chile
| | - Gonzalo A Jaña
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano 4260000, Chile
| | - José L Guzmán
- Laboratory of Molecular Neurobiology, Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepción 4070386, Chile
| | - Joel B Alderete
- Instituto de Química de Recursos Naturales, Universidad de Talca, Casilla 747, Talca, Chile
| | - Verónica A Jiménez
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano 4260000, Chile
| |
Collapse
|
65
|
Kumbhar BV, Bhandare VV. Exploring the interaction of Peloruside-A with drug resistant αβII and αβIII tubulin isotypes in human ovarian carcinoma using a molecular modeling approach. J Biomol Struct Dyn 2020; 39:1990-2002. [DOI: 10.1080/07391102.2020.1745689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Bajarang Vasant Kumbhar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | | |
Collapse
|
66
|
Discovery of isopenicin A, a meroterpenoid as a novel inhibitor of tubulin polymerization. Biochem Biophys Res Commun 2020; 525:303-307. [DOI: 10.1016/j.bbrc.2020.02.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/09/2023]
|
67
|
Estévez-Gallego J, Josa-Prado F, Ku S, Buey RM, Balaguer FA, Prota AE, Lucena-Agell D, Kamma-Lorger C, Yagi T, Iwamoto H, Duchesne L, Barasoain I, Steinmetz MO, Chrétien D, Kamimura S, Díaz JF, Oliva MA. Structural model for differential cap maturation at growing microtubule ends. eLife 2020; 9:50155. [PMID: 32151315 PMCID: PMC7064335 DOI: 10.7554/elife.50155] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/25/2020] [Indexed: 11/13/2022] Open
Abstract
Microtubules (MTs) are hollow cylinders made of tubulin, a GTPase responsible for essential functions during cell growth and division, and thus, key target for anti-tumor drugs. In MTs, GTP hydrolysis triggers structural changes in the lattice, which are responsible for interaction with regulatory factors. The stabilizing GTP-cap is a hallmark of MTs and the mechanism of the chemical-structural link between the GTP hydrolysis site and the MT lattice is a matter of debate. We have analyzed the structure of tubulin and MTs assembled in the presence of fluoride salts that mimic the GTP-bound and GDP•Pi transition states. Our results challenge current models because tubulin does not change axial length upon GTP hydrolysis. Moreover, analysis of the structure of MTs assembled in the presence of several nucleotide analogues and of taxol allows us to propose that previously described lattice expansion could be a post-hydrolysis stage involved in Pi release.
Collapse
Affiliation(s)
- Juan Estévez-Gallego
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Fernando Josa-Prado
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Siou Ku
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, Rennes, France
| | - Ruben M Buey
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain.,Departamento de Microbiología y Genética, Universidad de Salamanca-Campus Miguel de Unamuno, Salamanca, Spain
| | - Francisco A Balaguer
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Andrea E Prota
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen, Switzerland
| | - Daniel Lucena-Agell
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | | - Toshiki Yagi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Hiroyuki Iwamoto
- Diffraction and Scattering Division, Japan Synchrotron Radiation Research Institute, Hyogo, Japan
| | - Laurence Duchesne
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, Rennes, France
| | - Isabel Barasoain
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Michel O Steinmetz
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen, Switzerland.,University of Basel, Biozentrum, Basel, Switzerland
| | - Denis Chrétien
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, Rennes, France
| | - Shinji Kamimura
- Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - J Fernando Díaz
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Maria A Oliva
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| |
Collapse
|
68
|
Wang T, Wu C, Wang C, Zhang G, Arnst KE, Yao Y, Zhang Z, Wang Y, Pu D, Li W. Unraveling the molecular mechanism of BNC105, a phase II clinical trial vascular disrupting agent, provides insights into drug design. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30008-5. [PMID: 32085900 DOI: 10.1016/j.bbrc.2019.12.083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/08/2023]
Abstract
Microtubules are made up of tubulin protein and play a very important part in numerous cellular events of eukaryotic cells, which is why they are seen as attractive targets for tumor chemotherapy. BNC105, a known vascular targeting agent, has entered in phase II clinical trials. It has previously been confirmed that BNC105 is an effective microtubule targeting agent for various cancers. BNC105 exhibits selectivity for tumor cells, elicits vascular disrupting effects, and inhibits tumor growth. However, the molecular mechanism of BNC105 is still elusive. Herein, the crystal structure of BNC105 in complex with tubulin protein is revealed, demonstrating the its interaction with the colchicine binding site. In order to thoroughly evaluate its molecular mechanism from a structural-activity-relationship standpoint, the binding mode of tubulin to BNC-105 is compared with colchicine, CA-4 and other BNC-105 derivatives. Our study not only confirms the detailed interactions of the BNC105-tubulin complex, but also offer substantial structural foundation for the design and development of novel benzo[b]furan derivatives as microtubule targeting agents.
Collapse
Affiliation(s)
- Tao Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, PR China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, PR China
| | - Chengdi Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Guiping Zhang
- Bontac Bio-Engineering (Shenzhen) Co., Ltd, Shenzhen, Guangdong, 518102, PR China
| | - Kinsie E Arnst
- The University of Tennessee Health Science Center, Department of Pharmaceutical Sciences, Memphis, TN, 38163, United States
| | - Yijun Yao
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Zhixiong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, PR China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Dan Pu
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China.
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|
69
|
Niu L, Yang J, Yan W, Yu Y, Zheng Y, Ye H, Chen Q, Chen L. Reversible binding of the anticancer drug KXO1 (tirbanibulin) to the colchicine-binding site of β-tubulin explains KXO1's low clinical toxicity. J Biol Chem 2019; 294:18099-18108. [PMID: 31628188 DOI: 10.1074/jbc.ra119.010732] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/14/2019] [Indexed: 02/05/2023] Open
Abstract
KXO1 (tirbanibulin or KX2-391) is as a non-ATP-competitive inhibitor of SRC proto-oncogene nonreceptor tyrosine kinase (SRC) and is being clinically investigated for the management of various cancers and actinic keratosis. Recently, KXO1 has also been shown to strongly inhibit tubulin. Interestingly, unlike conventional tubulin-targeting drugs, KXO1 has exhibited low toxicity in preclinical and clinical studies, but the reason for this remains elusive, as are the KXO1-binding site and other details of the interaction of KXO1 with tubulin. Here, cell-based experiments revealed that KXO1 induces tubulin depolymerization and G2/M phase cell cycle arrest at low nanomolar concentrations, similar to colchicine, used as a positive control. Results from biochemical experiments, including an N,N-ethylenebis(iodoacetamide) competition assay, disclosed that KXO1 binds to the colchicine-binding site on β-tubulin, further confirmed by the crystal structure of the tubulin-KXO1 complex at 2.5-Å resolution. A high-quality electron density map of the crystallographic data enabled us to unambiguously determine the position and orientation of KXO1 in the colchicine-binding site, revealing the detailed interactions between KXO1 and tubulin. We also found that KXO1 binds reversibly to purified tubulin, induces a totally reversible cellular effect (G2/M cell cycle arrest), and possesses no cellular toxicity 5 days after drug washout, explaining KXO1's low toxicity. In summary, we show that KXO1 binds to the colchicine-binding site of tubulin and resolved the crystal structure of the tubulin-KXO1 complex. Importantly, KXO1's reversible binding to tubulin explains its clinically low toxicity, an insight that could guide further clinical applications of KXO1.
Collapse
Affiliation(s)
- Lu Niu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Yamei Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Yunhua Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611730, People's Republic of China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China.
| |
Collapse
|
70
|
Yang J, Yan W, Li Y, Niu L, Ye H, Chen L. The Natural Compound Withaferin A Covalently Binds to Cys239 of β-Tubulin to Promote Tubulin Degradation. Mol Pharmacol 2019; 96:711-719. [PMID: 31585985 DOI: 10.1124/mol.119.117812] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/28/2019] [Indexed: 02/05/2023] Open
Abstract
Withaferin A (WIT) is a natural product possessing a wide range of pharmacologic activities. Previous studies have reported covalent binding of WIT to tubulin and down-of tubulin protein levels although the underlying mechanisms remain to be established. In the current investigation, we showed that WIT induces down-regulation of tubulin in a post-transcriptional manner, suggestive of direct and potent activity in tubulin degradation. The N,N'-ethylene bis(iodoacetamide) assay and competitive binding experiments with four colchicine site-targeted tubulin inhibitors further revealed that WIT interacts with the colchicine site of tubulin to promote degradation. WIT irreversibly inhibited tubulin polymerization, and mass spectrometry results disclosed binding to cysteine at position 239 (Cys239) and Cys303 sites of β-tubulin. Interestingly, WIT promoted degradation of the β-tubulin isoforms containing Cys239 [β2, β4, and β5(β)] but had no effect on those containing Ser239 (β3 and β6). Moreover, a C239S but not C303S mutation in β-tubulin completely abolished the degradation effect of WIT, suggesting that the Cys239-WIT covalent bond accounts for this activity. Our collective results clearly demonstrate that covalent interactions between WIT and Cys239 of β-tubulin promote tubulin degradation, supporting its potential utility as a therapeutic compound. SIGNIFICANCE STATEMENT: Withaferin A, a natural product possessing a wide range of pharmacologic activities, covalently binds to Cys239 of β-tubulin near the colchicine site, and the WIT-Cys239 covalent bond accounts for WIT-induced tubulin degradation, fully clarifying the underlying mechanisms and supporting its potential utility a therapeutic compound.
Collapse
Affiliation(s)
- Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu, People's Republic of China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu, People's Republic of China
| | - Yong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu, People's Republic of China
| | - Lu Niu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu, People's Republic of China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu, People's Republic of China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu, People's Republic of China
| |
Collapse
|
71
|
An overview of microtubule targeting agents for cancer therapy. Arh Hig Rada Toksikol 2019; 70:160-172. [DOI: 10.2478/aiht-2019-70-3258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 09/01/2019] [Indexed: 12/27/2022] Open
Abstract
Abstract
The entire world is looking for effective cancer therapies whose benefits would outweigh their toxicity. One way to reduce resistance to chemotherapy and its adverse effects is the so called targeted therapy, which targets specific molecules (“molecular targets”) that play a critical role in cancer growth, progression, and metastasis. One such specific target are microtubules. In this review we address the current knowledge about microtubule-targeting agents or drugs (MTAs/MTDs) used in cancer therapy from their synthesis to toxicities. Synthetic and natural MTAs exhibit antitumor activity, and preclinical and clinical studies have shown that their anticancer effectiveness is higher than that of traditional drug therapies. Furthermore, MTAs involve a lower risk of adverse effects such as neurotoxicity and haemotoxicity. Several new generation MTAs are currently being evaluated for clinical use. This review brings updated information on the benefits of MTAs, therapeutic approaches, advantages, and challenges in their research.
Collapse
|
72
|
C SK, Gadewal N, Choudhary RK, Dasgupta D. Insights into the flexibility of the T3 loop and GTPase activating protein (GAP) domain of dimeric α and β tubulins from a molecular dynamics perspective. Comput Biol Chem 2019; 82:37-43. [PMID: 31255973 DOI: 10.1016/j.compbiolchem.2019.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/09/2019] [Accepted: 06/12/2019] [Indexed: 10/26/2022]
Abstract
Tubulin protein is the fundamental unit of microtubules, and comprises of α and β subunits arranged in an alternate manner forming protofilaments. These longitudinal protofilaments are made up of intra- (α-β) and inter-dimer (β-α) interactions. Literature review confirms that GTP hydrolysis results in considerable structural rearrangement within GTP binding site of β-α dimer interface after the release of γ phosphate. In addition to this, the intra-dimer interface exhibits structural rigidity which needs further investigation. In this study, we explored the reasons for the flexibility and the rigidity of the β-α dimer and the α-β dimer respectively through molecular simulation and Anisotropic Normal Mode based analysis. As per the sequence alignment report, two glycine residues (Gly96 and Gly98) were observed in the T3 loop of the β subunit which get substituted by Asp98 and Ala100 in the T3 loop of the α subunit. The higher mobility of glycine residues contributes to the flexibility of the T3 loop of inter-dimer when they come in direct contact with the GTPase Activating Protein (GAP) domain of the subunit. This was confirmed through RMSD, RMSF and Radius of Gyration based studies. Conversely, the intra-dimer exhibited a lower mobility in the absence of glycine residues. As per ANM based analysis, positive domain correlations were observed between T3 loop and GAP domain of intra- and inter- dimeric contact regions. However, these correlation motions were higher in the intra-dimer as compared to the inter-dimer interface. Thus on the basis of our findings, we hypothesize that the higher flexibility of T3 loop and the GAP domain of the inter-dimer is required for structural rearrangement and protofilament stability during hydrolysis. Furthermore, the slightly rigid nature of the T3 loop and the GAP domain of the intra-dimer assists in enhancing the monomer-monomer interaction through the higher positive domain correlation.
Collapse
Affiliation(s)
- Selvaa Kumar C
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India.
| | - Nikhil Gadewal
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Kharghar, Navi Mumbai, India.
| | - Rajan Kumar Choudhary
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Kharghar, Navi Mumbai, India
| | - Debjani Dasgupta
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai, India
| |
Collapse
|
73
|
Hanif N, Murni A, Tanaka C, Tanaka J. Marine Natural Products from Indonesian Waters. Mar Drugs 2019; 17:md17060364. [PMID: 31248122 PMCID: PMC6627775 DOI: 10.3390/md17060364] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
Natural products are primal and have been a driver in the evolution of organic chemistry and ultimately in science. The chemical structures obtained from marine organisms are diverse, reflecting biodiversity of genes, species and ecosystems. Biodiversity is an extraordinary feature of life and provides benefits to humanity while promoting the importance of environment conservation. This review covers the literature on marine natural products (MNPs) discovered in Indonesian waters published from January 1970 to December 2017, and includes 732 original MNPs, 4 structures isolated for the first time but known to be synthetic entities, 34 structural revisions, 9 artifacts, and 4 proposed MNPs. Indonesian MNPs were found in 270 papers from 94 species, 106 genera, 64 families, 32 orders, 14 classes, 10 phyla, and 5 kingdoms. The emphasis is placed on the structures of organic molecules (original and revised), relevant biological activities, structure elucidation, chemical ecology aspects, biosynthesis, and bioorganic studies. Through the synthesis of past and future data, huge and partly undescribed biodiversity of marine tropical invertebrates and their importance for crucial societal benefits should greatly be appreciated.
Collapse
Affiliation(s)
- Novriyandi Hanif
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University (Bogor Agricultural University), Bogor 16680, Indonesia.
| | - Anggia Murni
- Tropical Biopharmaca Research Center, IPB University (Bogor Agricultural University), Bogor 16128, Indonesia.
| | - Chiaki Tanaka
- Department of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Junichi Tanaka
- Department of Chemistry, Biology, and Marine Science, University of the Ryukyus, Nishihara, Okinawa 903-0213, Japan.
| |
Collapse
|
74
|
Sánchez-Murcia PA, Mills A, Cortés-Cabrera Á, Gago F. Unravelling the covalent binding of zampanolide and taccalonolide AJ to a minimalist representation of a human microtubule. J Comput Aided Mol Des 2019; 33:627-644. [DOI: 10.1007/s10822-019-00208-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/24/2019] [Indexed: 01/27/2023]
|
75
|
Revisiting microtubule targeting agents: α-Tubulin and the pironetin binding site as unexplored targets for cancer therapeutics. Bioorg Med Chem Lett 2019; 29:1865-1873. [PMID: 31130264 DOI: 10.1016/j.bmcl.2019.05.042] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/17/2019] [Accepted: 05/19/2019] [Indexed: 01/21/2023]
Abstract
Molecules that bind to tubulin and disrupt tubulin dynamics are known as microtubule targeting agents. Treatment with a microtubule targeting agent leads to cell cycle arrest followed by apoptosis. Tubulin inhibitors have been highly effective in the clinical treatment of a variety of tumors and are being investigated for treatment of several other diseases. Currently, all FDA approved microtubule inhibitors bind to β-tubulin. Given the overall success of tubulin-binding agents in anticancer chemotherapy, α-tubulin is an attractive and unexplored target. Herein, we will discuss pironetin, the only compound known to bind α-tubulin, with particular focus on the known biological properties, the total syntheses, exploration of its structure-activity relationship, and future directions.
Collapse
|
76
|
Guo H, Li X, Guo Y, Zhen L. An overview of tubulin modulators deposited in protein data bank. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02352-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
77
|
Zúñiga MA, Alderete JB, Jaña GA, Navarrete KR, Jiménez VA. Molecular modeling study on the differential microtubule‐stabilizing effect in singly‐ and doubly‐bonded complexes with peloruside A and paclitaxel. Proteins 2019; 87:668-678. [DOI: 10.1002/prot.25692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/15/2019] [Accepted: 04/04/2019] [Indexed: 01/26/2023]
Affiliation(s)
- Matías A. Zúñiga
- Department of Chemical Sciences, Faculty of Exact Sciencies, Universidad Andres BelloSede Concepción Autopista Concepción‐Talcahuano Talcahuano Chile
| | - Joel B. Alderete
- Instituto de Química de Recursos Naturales, Universidad de Talca Casilla Talca Chile
| | - Gonzalo A. Jaña
- Department of Chemical Sciences, Faculty of Exact Sciencies, Universidad Andres BelloSede Concepción Autopista Concepción‐Talcahuano Talcahuano Chile
| | - Karen R. Navarrete
- Department of Chemical Sciences, Faculty of Exact Sciencies, Universidad Andres BelloSede Concepción Autopista Concepción‐Talcahuano Talcahuano Chile
| | - Verónica A. Jiménez
- Department of Chemical Sciences, Faculty of Exact Sciencies, Universidad Andres BelloSede Concepción Autopista Concepción‐Talcahuano Talcahuano Chile
| |
Collapse
|
78
|
Yang J, Li Y, Yan W, Li W, Qiu Q, Ye H, Chen L. Covalent modification of Cys-239 in β-tubulin by small molecules as a strategy to promote tubulin heterodimer degradation. J Biol Chem 2019; 294:8161-8170. [PMID: 30940730 DOI: 10.1074/jbc.ra118.006325] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/23/2019] [Indexed: 02/05/2023] Open
Abstract
Clinical microtubule-targeting drugs are functionally divided into microtubule-destabilizing and microtubule-stabilizing agents. Drugs from both classes achieve microtubule inhibition by binding different sites on tubulin and inhibiting or promoting polymerization with no concomitant effects on the protein levels of tubulin heterodimers. Here, we have identified a series of small molecules with diverse structures potentially representing a third class of novel tubulin inhibitors that promote degradation by covalent binding to Cys-239 of β-tubulin. The small molecules highlighted in this study include T0070907 (a peroxisome proliferator-activated receptor γ inhibitor), T007-1 (a T0070907 derivative), T138067, N,N'-ethylene-bis(iodoacetamide) (EBI), and allyl isothiocyanate (AITC). Label-free quantitative proteomic analysis revealed that T007-1 promotes tubulin degradation with high selectivity. Mass spectrometry findings showed covalent binding of both T0070907 and T007-01 to Cys-239 of β-tubulin. Furthermore, T007-1 exerted a degradative effect on tubulin isoforms possessing Cys-239 (β2, β4, and β5(β)) but not those containing Ser-239 (β3, β6) or mutant β-tubulin with a C239S substitution. Three small molecules (T138067, EBI, and AITC) also reported to bind covalently to Cys-239 of β-tubulin similarly induced tubulin degradation. Our results strongly suggest that covalent modification of Cys-239 of β-tubulin by small molecules could serve as a novel strategy to promote tubulin heterodimer degradation. We propose that these small molecules represent a third novel class of tubulin inhibitor agents that exert their effects through degradation activity.
Collapse
Affiliation(s)
- Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weimin Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
79
|
Balaguer FDA, Mühlethaler T, Estévez-Gallego J, Calvo E, Giménez-Abián JF, Risinger AL, Sorensen EJ, Vanderwal CD, Altmann KH, Mooberry SL, Steinmetz MO, Oliva MÁ, Prota AE, Díaz JF. Crystal Structure of the Cyclostreptin-Tubulin Adduct: Implications for Tubulin Activation by Taxane-Site Ligands. Int J Mol Sci 2019; 20:ijms20061392. [PMID: 30897704 PMCID: PMC6471726 DOI: 10.3390/ijms20061392] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 12/22/2022] Open
Abstract
It has been proposed that one of the mechanisms of taxane-site ligand-mediated tubulin activation is modulation of the structure of a switch element (the M-loop) from a disordered form in dimeric tubulin to a folded helical structure in microtubules. Here, we used covalent taxane-site ligands, including cyclostreptin, to gain further insight into this mechanism. The crystal structure of cyclostreptin-bound tubulin reveals covalent binding to βHis229, but no stabilization of the M-loop. The capacity of cyclostreptin to induce microtubule assembly compared to other covalent taxane-site agents demonstrates that the induction of tubulin assembly is not strictly dependent on M-loop stabilization. We further demonstrate that most covalent taxane-site ligands are able to partially overcome drug resistance mediated by βIII-tubulin (βIII) overexpression in HeLa cells, and compare their activities to pironetin, an interfacial covalent inhibitor of tubulin assembly that displays invariant growth inhibition in these cells. Our findings suggest a relationship between a diminished interaction of taxane-site ligands with βIII-tubulin and βIII tubulin-mediated drug resistance. This supports the idea that overexpression of βIII increases microtubule dynamicity by counteracting the enhanced microtubule stability promoted by covalent taxane-site binding ligands.
Collapse
Affiliation(s)
- Francisco de Asís Balaguer
- Structural and Chemical Biology Department. Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Tobias Mühlethaler
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland.
| | - Juan Estévez-Gallego
- Structural and Chemical Biology Department. Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Enrique Calvo
- Unidad de Proteómica. Centro Nacional de Investigaciones Cardiovasculares, CNIC. Melchor Fernández de Almagro 3, 28029 Madrid, Spain.
| | - Juan Francisco Giménez-Abián
- Structural and Chemical Biology Department. Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - April L Risinger
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA.
| | - Erik J Sorensen
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA.
| | - Christopher D Vanderwal
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, CA 92697-2025, USA.
| | - Karl-Heinz Altmann
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zürich, Switzerland.
| | - Susan L Mooberry
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA.
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland.
- University of Basel, Biozentrum, 4056 Basel, Switzerland.
| | - María Ángela Oliva
- Structural and Chemical Biology Department. Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland.
| | - J Fernando Díaz
- Structural and Chemical Biology Department. Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
80
|
Chany AC, Legros F, Haroun H, Kundu UK, Biletskyi B, Torlak S, Mathé-Allainmat M, Lebreton J, Macé A, Carboni B, Renoux B, Gosselin P, Dujardin G, Gaulon-Nourry C. Function-Oriented Synthesis toward Peloruside A Analogues. Org Lett 2019; 21:2988-2992. [PMID: 30859834 DOI: 10.1021/acs.orglett.9b00413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A convergent and rapid synthesis of original C2,C3-unsaturated, C11,C13-keto-enol macrocycles with a peloruside A skeleton has been developed. These original unsaturated macrocycles constitute valuable platforms to access peloruside A analogues with high diversity. The four-fragment strategy implemented features two aldol-type couplings with the central C12-C14 building block TES-diazoacetone and a late-stage ring-closing metathesis. Enantiopure analogue 18ab showed antiproliferative activity in the low micromolar range on NCI and MCF7 tumor cell lines.
Collapse
Affiliation(s)
- Anne-Caroline Chany
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Frédéric Legros
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Heloua Haroun
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Uday Kumar Kundu
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Bohdan Biletskyi
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Sergii Torlak
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Monique Mathé-Allainmat
- Université de Nantes , Laboratoire CEISAM, UMR 6230 CNRS, Faculté des Sciences et des Techniques , Nantes , 44322 Cedex 3 , France
| | - Jacques Lebreton
- Université de Nantes , Laboratoire CEISAM, UMR 6230 CNRS, Faculté des Sciences et des Techniques , Nantes , 44322 Cedex 3 , France
| | - Aurélie Macé
- Univ Rennes, CNRS, ISCR [(Institut des Sciences Chimiques de Rennes)], UMR 6226 , F-35000 Rennes , France
| | - Bertrand Carboni
- Univ Rennes, CNRS, ISCR [(Institut des Sciences Chimiques de Rennes)], UMR 6226 , F-35000 Rennes , France
| | - Brigitte Renoux
- Institut de Chimie des Milieux et des Matériaux de Poitiers, IC2MP , Université de Poitiers, UMR 7285 CNRS , 4 rue Michel Brunet , 86022 Poitiers , France
| | - Pascal Gosselin
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Gilles Dujardin
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| | - Catherine Gaulon-Nourry
- Institut des Molécules et Matériaux , IMMM UMR 6283 CNRS - Le Mans Université , Avenue Olivier Messiaen , Le Mans 72085 Cedex 9 , France
| |
Collapse
|
81
|
Arnst KE, Banerjee S, Chen H, Deng S, Hwang DJ, Li W, Miller DD. Current advances of tubulin inhibitors as dual acting small molecules for cancer therapy. Med Res Rev 2019; 39:1398-1426. [PMID: 30746734 DOI: 10.1002/med.21568] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 12/25/2022]
Abstract
Microtubule (MT)-targeting agents are highly successful drugs as chemotherapeutic agents, and this is attributed to their ability to target MT dynamics and interfere with critical cellular functions, including, mitosis, cell signaling, intracellular trafficking, and angiogenesis. Because MT dynamics vary in the different stages of the cell cycle, these drugs tend to be the most effective against mitotic cells. While this class of drug has proven to be effective against many cancer types, significant hurdles still exist and include overcoming aspects such as dose limited toxicities and the development of resistance. Newer generations of developed drugs attack these problems and alternative approaches such as the development of dual tubulin and kinase inhibitors are being investigated. This approach offers the potential to show increased efficacy and lower toxicities. This review covers different categories of MT-targeting agents, recent advances in dual inhibitors, and current challenges for this drug target.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
82
|
Majumdar S, Basu D, Ghosh Dastidar S. Conformational States of E7010 Is Complemented by Microclusters of Water Inside the α,β-Tubulin Core. J Chem Inf Model 2018; 59:2274-2286. [PMID: 30516382 DOI: 10.1021/acs.jcim.8b00538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The α,β-tubulin is the building block of microtubules, which is associated with and dissociated from the microtubular architecture complying with the dynamic instability of the microtubules. This dynamic instability has a direct relation with the spindle formation by the microtubules and cell division kinetics. E7010 is one of the promising ligands of an α,β-tubulin protein that binds at the core of this protein and can diminish the protein's ability to fit to a growing microtubule, thus frustrating cell division. Although X-ray crystallography has reported a specific binding conformation of E7010 in PDB, molecular dynamics (MD) simulations have revealed two other conformational states of the ligand capable of binding to tubulin with stabilities close to that state reported in PDB. To rationalize this quasidegeneracy of ligand binding modes, MD simulations have further revealed that the understanding of the mechanism of E7010-tubulin binding remains incomplete unless the role of water molecules to bridge this interaction is taken into consideration, a very critical insight that was not visible from the PDB structure. Further, these water molecules differ from the standard examples of "bridging" waters which generally exist as isolated water molecules between the receptor and the ligand. In the present case, the water molecules sandwiched between ligand and protein, sequestered from the bulk solvent, integrate with each other by an H-bonds network forming a group, which appear as microclusters of water. The structural packing with the ligand binding pocket and the bridging interactions between protein and ligand take place through such clusters. The presence of this microcluster of water is not just cosmetic, instead they have a crucial impact on the ligand binding thermodynamics. Only with the explicit consideration of these water clusters in the binding energy calculations (MMGBSA) is the stability of the native mode of ligand binding reported in PDB rationalized. At the same time, two other binding modes are elucidated to be quasi-degenerate with the native state and that indicates the further possibility in gaining more entropic stabilization of the complex. The role of such "bridging" water clusters to enhance the protein-ligand interaction will be insightful for designing the next generation prospective compounds in the field of cancer therapeutics.
Collapse
Affiliation(s)
- Sarmistha Majumdar
- Division of Bioinformatics , Bose Institute , P-1/12 C.I.T. Scheme VII M , Kolkata 700054 , India
| | - Debadrita Basu
- Division of Bioinformatics , Bose Institute , P-1/12 C.I.T. Scheme VII M , Kolkata 700054 , India
| | - Shubhra Ghosh Dastidar
- Division of Bioinformatics , Bose Institute , P-1/12 C.I.T. Scheme VII M , Kolkata 700054 , India
| |
Collapse
|
83
|
Vicente-Blázquez A, González M, Álvarez R, Del Mazo S, Medarde M, Peláez R. Antitubulin sulfonamides: The successful combination of an established drug class and a multifaceted target. Med Res Rev 2018; 39:775-830. [PMID: 30362234 DOI: 10.1002/med.21541] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/02/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
Tubulin, the microtubules and their dynamic behavior are amongst the most successful antitumor, antifungal, antiparasitic, and herbicidal drug targets. Sulfonamides are exemplary drugs with applications in the clinic, in veterinary and in the agrochemical industry. This review summarizes the actual state and recent progress of both fields looking from the double point of view of the target and its drugs, with special focus onto the structural aspects. The article starts with a brief description of tubulin structure and its dynamic assembly and disassembly into microtubules and other polymers. Posttranslational modifications and the many cellular means of regulating and modulating tubulin's biology are briefly presented in the tubulin code. Next, the structurally characterized drug binding sites, their occupying drugs and the effects they induce are described, emphasizing on the structural requirements for high potency, selectivity, and low toxicity. The second part starts with a summary of the favorable and highly tunable combination of physical-chemical and biological properties that render sulfonamides a prototypical example of privileged scaffolds with representatives in many therapeutic areas. A complete description of tubulin-binding sulfonamides is provided, covering the different species and drug sites. Some of the antimitotic sulfonamides have met with very successful applications and others less so, thus illustrating the advances, limitations, and future perspectives of the field. All of them combine in a mechanism of action and a clinical outcome that conform efficient drugs.
Collapse
Affiliation(s)
- Alba Vicente-Blázquez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Myriam González
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Sara Del Mazo
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Manuel Medarde
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
84
|
Hong SW, Singh AJ, Patel V, Russell ER, Field JJ, Miller JH, Northcote PT. Peloruside E (22-Norpeloruside A), a Pelorusane Macrolide from the New Zealand Marine Sponge Mycale hentscheli, Retains Microtubule-Stabilizing Properties. JOURNAL OF NATURAL PRODUCTS 2018; 81:2125-2128. [PMID: 30188708 DOI: 10.1021/acs.jnatprod.8b00557] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A new peloruside congener, peloruside E (5), has been isolated in sub-milligram quantities from a specimen of the New Zealand marine sponge Mycale hentscheli. The structure of 5 differs from the parent compound peloruside A (1) by replacement of the C-10 gem-dimethyl moiety with a monomethyl substituent and represents the first structural deviation in the pelorusane scaffold. Peloruside E (5) is potently antiproliferative (HL-60, IC50 90 nM, cf. 1, 19 nM) and polymerizes purified tubulin, albeit at a rate lower than that of 1.
Collapse
Affiliation(s)
- Sa Weon Hong
- School of Chemical and Physical Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - A Jonathan Singh
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
- Ferrier Research Institute , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - Vimal Patel
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
- School of Biological Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - Euan R Russell
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
- School of Biological Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - Jessica J Field
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
- School of Biological Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - John H Miller
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
- School of Biological Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - Peter T Northcote
- Centre for Biodiscovery , Victoria University of Wellington , Wellington 6012 , New Zealand
- Ferrier Research Institute , Victoria University of Wellington , Wellington 6012 , New Zealand
| |
Collapse
|
85
|
Microtubule-Targeting Agents: Strategies To Hijack the Cytoskeleton. Trends Cell Biol 2018; 28:776-792. [PMID: 29871823 DOI: 10.1016/j.tcb.2018.05.001] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 11/20/2022]
Abstract
Microtubule-targeting agents (MTAs) such as paclitaxel and the vinca alkaloids are among the most important medical weapons available to combat cancer. MTAs interfere with intracellular transport, inhibit eukaryotic cell proliferation, and promote cell death by suppressing microtubule dynamics. Recent advances in the structural analysis of MTAs have enabled the extensive characterization of their interactions with microtubules and their building block tubulin. We review here our current knowledge on the molecular mechanisms used by MTAs to hijack the microtubule cytoskeleton, and discuss dual inhibitors that target both kinases and microtubules. We further formulate some outstanding questions related to MTA structural biology and present possible routes for future investigations of this fascinating class of antimitotic agents.
Collapse
|
86
|
Yang J, Yan W, Yu Y, Wang Y, Yang T, Xue L, Yuan X, Long C, Liu Z, Chen X, Hu M, Zheng L, Qiu Q, Pei H, Li D, Wang F, Bai P, Wen J, Ye H, Chen L. The compound millepachine and its derivatives inhibit tubulin polymerization by irreversibly binding to the colchicine-binding site in β-tubulin. J Biol Chem 2018; 293:9461-9472. [PMID: 29691282 DOI: 10.1074/jbc.ra117.001658] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/19/2018] [Indexed: 02/05/2023] Open
Abstract
Inhibitors that bind to the paclitaxel- or vinblastine-binding sites of tubulin have been part of the pharmacopoeia of anticancer therapy for decades. However, tubulin inhibitors that bind to the colchicine-binding site are not used in clinical cancer therapy, because of their low therapeutic index. To address multidrug resistance to many conventional tubulin-binding agents, numerous efforts have attempted to clinically develop inhibitors that bind the colchicine-binding site. Previously, we have found that millepachine (MIL), a natural chalcone-type small molecule extracted from the plant Millettia pachycarpa, and its two derivatives (MDs) SKLB028 and SKLB050 have potential antitumor activities both in vitro and in vivo However, their cellular targets and mechanisms are unclear. Here, biochemical and cellular experiments revealed that the MDs directly and irreversibly bind β-tubulin. X-ray crystallography of the tubulin-MD structures disclosed that the MDs bind at the tubulin intradimer interface and to the same site as colchicine and that their binding mode is similar to that of colchicine. Of note, MDs inhibited tubulin polymerization and caused G2/M cell-cycle arrest. Comprehensive analysis further revealed that free MIL exhibits an s-cis conformation, whereas MIL in the colchicine-binding site in tubulin adopts an s-trans conformation. Moreover, introducing an α-methyl to MDs to increase the proportion of s-trans conformations augmented MDs' tubulin inhibition activity. Our study uncovers a new class of chalcone-type tubulin inhibitors that bind the colchicine-binding site in β-tubulin and suggests that the s-trans conformation of these compounds may make them more active anticancer agents.
Collapse
Affiliation(s)
- Jianhong Yang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Wei Yan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Yamei Yu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Yuxi Wang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Tao Yang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Linlin Xue
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Xue Yuan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Caofeng Long
- the Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523325, China
| | - Zuowei Liu
- the Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523325, China
| | - Xiaoxin Chen
- the Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523325, China
| | - Mengshi Hu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Li Zheng
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Qiang Qiu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Heying Pei
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Dan Li
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Fang Wang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Peng Bai
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Jiaolin Wen
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Haoyu Ye
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Lijuan Chen
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| |
Collapse
|
87
|
Xiang W, Choudhary S, Hamel E, Mooberry SL, Gangjee A. Structure based drug design and in vitro metabolism study: Discovery of N-(4-methylthiophenyl)-N,2-dimethyl-cyclopenta[d]pyrimidine as a potent microtubule targeting agent. Bioorg Med Chem 2018; 26:2437-2451. [PMID: 29655610 DOI: 10.1016/j.bmc.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 03/26/2018] [Accepted: 04/03/2018] [Indexed: 01/07/2023]
Abstract
We report a series of tubulin targeting agents, some of which demonstrate potent antiproliferative activities. These analogs were designed to optimize the antiproliferative activity of 1 by varying the heteroatom substituent at the 4'-position, the basicity of the 4-position amino moiety, and conformational restriction. The potential metabolites of the active compounds were also synthesized. Some compounds demonstrated single digit nanomolar IC50 values for antiproliferative effects in MDA-MB-435 melanoma cells. Particularly, the S-methyl analog 3 was more potent than 1 in MDA-MB-435 cells (IC50 = 4.6 nM). Incubation of 3 with human liver microsomes showed that the primary metabolite of the S-methyl moiety of 3 was the methyl sulfinyl group, as in analog 5. This metabolite was equipotent with the lead compound 1 in MDA-MB-435 cells (IC50 = 7.9 nM). Molecular modeling and electrostatic surface area were determined to explain the activities of the analogs. Most of the potent compounds overcome multiple mechanisms of drug resistance and compound 3 emerged as the lead compound for further SAR and preclinical development.
Collapse
Affiliation(s)
- Weiguo Xiang
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Shruti Choudhary
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Susan L Mooberry
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States.
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States.
| |
Collapse
|
88
|
Miller JH, Field JJ, Kanakkanthara A, Owen JG, Singh AJ, Northcote PT. Marine Invertebrate Natural Products that Target Microtubules. JOURNAL OF NATURAL PRODUCTS 2018; 81:691-702. [PMID: 29431439 DOI: 10.1021/acs.jnatprod.7b00964] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Marine natural products as secondary metabolites are a potential major source of new drugs for treating disease. In some cases, cytotoxic marine metabolites target the microtubules of the eukaryote cytoskeleton for reasons that will be discussed. This review covers the microtubule-targeting agents reported from sponges, corals, tunicates, and molluscs and the evidence that many of these secondary metabolites are produced by bacterial symbionts. The review finishes by discussing the directions for future development and production of clinically relevant amounts of these natural products and their analogues through aquaculture, chemical synthesis, and biosynthesis by bacterial symbionts.
Collapse
Affiliation(s)
| | | | - Arun Kanakkanthara
- Department of Oncology and Department of Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , Minnesota , United States
| | | | | | | |
Collapse
|
89
|
Field JJ, Pera B, Gallego JE, Calvo E, Rodríguez-Salarichs J, Sáez-Calvo G, Zuwerra D, Jordi M, Andreu JM, Prota AE, Ménchon G, Miller JH, Altmann KH, Díaz JF. Zampanolide Binding to Tubulin Indicates Cross-Talk of Taxane Site with Colchicine and Nucleotide Sites. JOURNAL OF NATURAL PRODUCTS 2018; 81:494-505. [PMID: 29023132 DOI: 10.1021/acs.jnatprod.7b00704] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The marine natural product zampanolide and analogues thereof constitute a new chemotype of taxoid site microtubule-stabilizing agents with a covalent mechanism of action. Zampanolide-ligated tubulin has the switch-activation loop (M-loop) in the assembly prone form and, thus, represents an assembly activated state of the protein. In this study, we have characterized the biochemical properties of the covalently modified, activated tubulin dimer, and we have determined the effect of zampanolide on tubulin association and the binding of tubulin ligands at other binding sites. Tubulin activation by zampanolide does not affect its longitudinal oligomerization but does alter its lateral association properties. The covalent binding of zampanolide to β-tubulin affects both the colchicine site, causing a change of the quantum yield of the bound ligand, and the exchangeable nucleotide binding site, reducing the affinity for the nucleotide. While these global effects do not change the binding affinity of 2-methoxy-5-(2,3,4-trimethoxyphenyl)-2,4,6-cycloheptatrien-1-one (MTC) (a reversible binder of the colchicine site), the binding affinity of a fluorescent analogue of GTP (Mant-GTP) at the nucleotide E-site is reduced from 12 ± 2 × 105 M-1 in the case of unmodified tubulin to 1.4 ± 0.3 × 105 M-1 in the case of the zampanolide tubulin adduct, indicating signal transmission between the taxane site and the colchicine and nucleotide sites of β-tubulin.
Collapse
Affiliation(s)
- Jessica J Field
- Centre for Biodiscovery, School of Biological Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - Benet Pera
- Centro de Investigaciones Biológicas (CIB) , CSIC , Madrid 28048 , Spain
| | | | - Enrique Calvo
- Unidad de Proteómica , Centro Nacional de Investigaciones Cardiovasculares , Madrid 28029 , Spain
| | | | - Gonzalo Sáez-Calvo
- Centro de Investigaciones Biológicas (CIB) , CSIC , Madrid 28048 , Spain
| | - Didier Zuwerra
- Department of Chemistry and Applied Biosciences , Swiss Federal Institute of Technology (ETH) Zürich, Institute of Pharmaceutical Sciences , HCI H405, Zürich 8092 , Switzerland
| | - Michel Jordi
- Department of Chemistry and Applied Biosciences , Swiss Federal Institute of Technology (ETH) Zürich, Institute of Pharmaceutical Sciences , HCI H405, Zürich 8092 , Switzerland
| | - José M Andreu
- Centro de Investigaciones Biológicas (CIB) , CSIC , Madrid 28048 , Spain
| | - Andrea E Prota
- Laboratory of Biomolecular Research , Paul Scherrer Institut , Villigen PSI 5232 , Switzerland
| | - Grégory Ménchon
- Laboratory of Biomolecular Research , Paul Scherrer Institut , Villigen PSI 5232 , Switzerland
| | - John H Miller
- Centre for Biodiscovery, School of Biological Sciences , Victoria University of Wellington , Wellington 6012 , New Zealand
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences , Swiss Federal Institute of Technology (ETH) Zürich, Institute of Pharmaceutical Sciences , HCI H405, Zürich 8092 , Switzerland
| | - J Fernando Díaz
- Centro de Investigaciones Biológicas (CIB) , CSIC , Madrid 28048 , Spain
| |
Collapse
|
90
|
Castro-Alvarez A, Pineda O, Vilarrasa J. Further Insight into the Interactions of the Cytotoxic Macrolides Laulimalide and Peloruside A with Their Common Binding Site. ACS OMEGA 2018; 3:1770-1782. [PMID: 31458493 PMCID: PMC6641392 DOI: 10.1021/acsomega.7b01723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 01/23/2018] [Indexed: 06/10/2023]
Abstract
The binding site of the macrolides laulimalide and peloruside A, which is different from that of the clinically useful drugs paclitaxel/taxol and ixabepilone (tax site), is known to be between two adjacent β-tubulin units (ext site). Here, we report our study of the binding of these molecules to an α1β1/α2β2-tubulin "tetramer" model. AutoDock 4.2.6//AutoDock Vina dockings predicted that the affinities of laulimalide and peloruside A for the tax site are quite similar to those for the ext site. However, molecular dynamics (MD) simulations indicated that only when these two ligands are located at the ext site, there are contacts that help stabilize the system, favoring the β1/β2 interactions. The binding affinity of laulimalide for this site is stronger than that of peloruside A, but this is compensated for by additional β1/β2 contacts that are induced by peloruside A. MD studies also suggested that epothilones at the tax site and either laulimalide or peloruside A at the ext site cause similar stabilizing effects (mainly linking the M-loop of β1 and loop H1-B2 of β2). In a "hexamer" model (3 units of αβ-tubulin), the effects are confirmed. Metadynamics simulations of laulimalide and peloruside A, which are reported for the first time, suggest that peloruside A produces a stronger change in the M-loop, which explains the stabilization of the β1/β2 interaction.
Collapse
|
91
|
Zúñiga MA, Alderete JB, Jaña GA, Fernandez PA, Ramos MJ, Jiménez VA. Modulation of lateral and longitudinal interdimeric interactions in microtubule models by Laulimalide and Peloruside A association: A molecular modeling approach on the mechanism of microtubule stabilizing agents. Chem Biol Drug Des 2018; 91:1042-1055. [DOI: 10.1111/cbdd.13168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/27/2017] [Accepted: 12/31/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Matías A. Zúñiga
- Departamento de Ciencias Químicas; Facultad de Ciencias Exactas; Universidad Andres Bello; Talcahuano Chile
| | - Joel B. Alderete
- Departamento de Química Orgánica; Facultad de Ciencias Químicas; Universidad de Concepción; Concepción Chile
| | - Gonzalo A. Jaña
- Departamento de Ciencias Químicas; Facultad de Ciencias Exactas; Universidad Andres Bello; Talcahuano Chile
| | | | - Maria J. Ramos
- Faculdade de Ciencias; Universidad do Porto; Porto Portugal
| | - Verónica A. Jiménez
- Departamento de Ciencias Químicas; Facultad de Ciencias Exactas; Universidad Andres Bello; Talcahuano Chile
| |
Collapse
|
92
|
Guzmán-Ocampo DC, Aguayo-Ortiz R, Cano-González L, Castillo R, Hernández-Campos A, Dominguez L. Effects of the Protonation State of Titratable Residues and the Presence of Water Molecules on Nocodazole Binding to β-Tubulin. ChemMedChem 2017; 13:20-24. [PMID: 29059502 DOI: 10.1002/cmdc.201700530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/18/2017] [Indexed: 01/18/2023]
Abstract
Regulation of microtubule assembly by antimitotic agents is a potential therapeutic strategy for the treatment of cancer, parasite infections, and neurodegenerative diseases. One of these agents is nocodazole (NZ), which inhibits microtubule polymerization by binding to β-tubulin. NZ was recently co-crystallized in Gallus gallus tubulin, providing new information about the features of interaction for ligand recognition and stability. In this work, we used state-of-the-art computational approaches to evaluate the protonation effects of titratable residues and the presence of water molecules in the binding of NZ. Analysis of protonation states showed that residue E198 has the largest modification in its pKa value. The resulting E198 pKa value, calculated with pH-REMD methodology (pKa =6.21), was higher than the isolated E amino acid (pKa =4.25), thus being more likely to be found in its protonated state at the binding site. Moreover, we identified an interaction between a water molecule and C239 and G235 as essential for NZ binding. Our results suggest that the protonation state of E198 and the structural water molecules play key roles in the binding of NZ to β-tubulin.
Collapse
Affiliation(s)
- Dulce C Guzmán-Ocampo
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Rodrigo Aguayo-Ortiz
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Lucia Cano-González
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Rafael Castillo
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Alicia Hernández-Campos
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Laura Dominguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| |
Collapse
|
93
|
Dicitrinone D, an antimitotic polyketide isolated from the marine-derived fungus Penicillium citrinum. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.08.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
94
|
Niu L, Wang Y, Wang C, Wang Y, Jiang X, Ma L, Wu C, Yu Y, Chen Q. Structure of 4'-demethylepipodophyllotoxin in complex with tubulin provides a rationale for drug design. Biochem Biophys Res Commun 2017; 493:718-722. [PMID: 28864414 DOI: 10.1016/j.bbrc.2017.08.125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/28/2017] [Indexed: 02/05/2023]
Abstract
Microtubules consists of αβ-tubulin heterodimers and are highly attractive targets for anti-cancer drugs. A broad range of agents have been identified to bind to tubulin and interfere with microtubule assembly, including colchicine binding site inhibitors (CBSIs). Podophyllotoxin is a CBSI that inhibits the assembly of microtubules. However, for a long time, the design and development of podophyllotoxin family drugs have been hindered by the lack of high-resolution structural information of the tubulin-agent complex. We report the first high-resolution (2.8 Å) structure of a podophyllotoxin family agent (4'-demethylepipodophyllotoxin, DMEP) complexed with tubulin and revealed the detailed interactions between DMEP and tubulin. Comparison of this structure and other CBSIs explains previous results of the structure-activity-relationship (SAR) studies, and provides insights into the development of new podophyllotoxin derivatives targeting the colchicine site.
Collapse
Affiliation(s)
- Lu Niu
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China
| | - Yuxi Wang
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China; Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guo Xue Street, Chengdu, Sichuan, 610041, PR China
| | - Chengdi Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guo Xue Street, Chengdu, Sichuan, 610041, PR China
| | - Yanyan Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guo Xue Street, Chengdu, Sichuan, 610041, PR China; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guo Xue Street, Chengdu, Sichuan, 610041, PR China
| | - Xiaohua Jiang
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China
| | - Lingling Ma
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China
| | - Chengyong Wu
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China
| | - Yamei Yu
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China.
| | - Qiang Chen
- Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, PR China.
| |
Collapse
|
95
|
Yang CPH, Horwitz SB. Taxol ®: The First Microtubule Stabilizing Agent. Int J Mol Sci 2017; 18:ijms18081733. [PMID: 28792473 PMCID: PMC5578123 DOI: 10.3390/ijms18081733] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022] Open
Abstract
Taxol®, an antitumor drug with significant activity, is the first microtubule stabilizing agent described in the literature. This short review of the mechanism of action of Taxol® emphasizes the research done in the Horwitz’ laboratory. It discusses the contribution of photoaffinity labeled analogues of Taxol® toward our understanding of the binding site of the drug on the microtubule. The importance of hydrogen/deuterium exchange experiments to further our insights into the stabilization of microtubules by Taxol® is addressed. The development of drug resistance, a major problem that arises in the clinic, is discussed. Studies describing differential drug binding to distinct β-tubulin isotypes are presented. Looking forward, it is suggested that the β-tubulin isotype content of a tumor may influence its responses to Taxol®.
Collapse
Affiliation(s)
- Chia-Ping Huang Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Department of Obstetrics and Gynecology and Women's Health, Division of Gynecologic Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Susan Band Horwitz
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
96
|
Tubulin Inhibitor-Based Antibody-Drug Conjugates for Cancer Therapy. Molecules 2017; 22:molecules22081281. [PMID: 28763044 PMCID: PMC6152078 DOI: 10.3390/molecules22081281] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/29/2017] [Indexed: 11/16/2022] Open
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. 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.
Collapse
|
97
|
Shaik SP, Vishnuvardhan M, Sultana F, Subba Rao A, Bagul C, Bhattacharjee D, Kapure JS, Jain N, Kamal A. Design and synthesis of 1,2,3-triazolo linked benzo[ d ]imidazo[2,1- b ]thiazole conjugates as tubulin polymerization inhibitors. Bioorg Med Chem 2017; 25:3285-3297. [DOI: 10.1016/j.bmc.2017.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/11/2017] [Indexed: 12/23/2022]
|
98
|
Quinolin-6-Yloxyacetamides Are Microtubule Destabilizing Agents That Bind to the Colchicine Site of Tubulin. Int J Mol Sci 2017. [PMID: 28640209 PMCID: PMC5535829 DOI: 10.3390/ijms18071336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Quinolin-6-yloxyacetamides (QAs) are a chemical class of tubulin polymerization inhibitors that were initially identified as fungicides. Here, we report that QAs are potent anti-proliferative agents against human cancer cells including ones that are drug-resistant. QAs act by disrupting the microtubule cytoskeleton and by causing severe mitotic defects. We further demonstrate that QAs inhibit tubulin polymerization in vitro. The high resolution crystal structure of the tubulin-QA complex revealed that QAs bind to the colchicine site on tubulin, which is targeted by microtubule-destabilizing agents such as colchicine and nocodazole. Together, our data establish QAs as colchicine-site ligands and explain the molecular mechanism of microtubule destabilization by this class of compounds. They further extend our structural knowledge on antitubulin agents and thus should aid in the development of new strategies for the rational design of ligands against multidrug-resistant cancer cells.
Collapse
|
99
|
Nogales E, Kellogg EH. Challenges and opportunities in the high-resolution cryo-EM visualization of microtubules and their binding partners. Curr Opin Struct Biol 2017. [PMID: 28628789 DOI: 10.1016/j.sbi.2017.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
As non-crystallizable polymers, microtubules have been the target of cryo-electron microscopy (cryo-EM) studies since the technique was first established. Over the years, image processing strategies have been developed that take care of the unique, pseudo-helical symmetry of the microtubule. With recent progress in data quality and data processing, cryo-EM reconstructions are now reaching resolutions that allow the generation of atomic models of microtubules and the factors that bind them. These include cellular partners that contribute to microtubule cellular functions, or small ligands that interfere with those functions in the treatment of cancer. The stage is set to generate a family portrait for all identified microtubule interacting proteins and to use cryo-EM as a drug development tool in the targeting of tubulin.
Collapse
Affiliation(s)
- Eva Nogales
- Molecular and Cell Biology Department and QB3 Institute, University of California Berkeley, CA 94720-3220, United States; Howard Hughes Medical Institute, University of California Berkeley, CA 94720-3220, United States; Molecular Biophysics and Integrative Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States.
| | - Elizabeth H Kellogg
- Molecular Biophysics and Integrative Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| |
Collapse
|
100
|
Zúñiga MA, Alderete JB, Jaña GA, Jiménez VA. Structural insight into the role of Gln293Met mutation on the Peloruside A/Laulimalide association with αβ-tubulin from molecular dynamics simulations, binding free energy calculations and weak interactions analysis. J Comput Aided Mol Des 2017; 31:643-652. [DOI: 10.1007/s10822-017-0029-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/28/2017] [Indexed: 01/27/2023]
|