1
|
Chamariya R, Suvarna V. Role of KSP inhibitors as anti-cancer therapeutics: an update. Anticancer Agents Med Chem 2022; 22:2517-2538. [PMID: 35043768 DOI: 10.2174/1871520622666220119093105] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/03/2021] [Accepted: 12/02/2021] [Indexed: 11/22/2022]
Abstract
Regardless of the growing discovery of anticancer treatments, targeting cancer-specific pathways, cytotoxic therapy still maintained its abundant clinical significance based on the fact that tumours harbour a greater population of actively dividing cells than normal tissues. Conventional anti-mitotic agents or microtubule poisons acting on the major mitotic spindle protein tubulin have been effectively used in clinical settings for cancer chemotherapy over the last three decades. However, use of these drugs is associated with limited clinical utility due to serious side effects such as debilitating and dose-limiting peripheral neuropathy, myelosuppression, drug resistance and allergic reactions. Therefore, research initiatives have been undertaken to develop novel microtubule motor proteins inhibitors that can potentially circumvent the limitations associated with conventional microtubule poisons. Kinesin spindle proteins (KSP) belonging to the kinesin-5 family play a crucial role during mitosis and unregulated cell proliferation. Several evidences from preclinical studies and different phases of clinical trials have presented kinesin spindle protein as a promising target for cancer therapeutics. kinesin spindle protein inhibitors causing mitosis disruption without interfering with microtubule dynamics in non-dividing cells offer a potential therapeutic alternative for the management of several major cancer types and are devoid of side effects associated with classical anti-mitotic drugs. This review summarizes recent data highlighting progress in the discovery of targeted KSP inhibitors and presents the development of scaffolds, structure-activity relationships, and outcomes of biological, and enzyme inhibition studies. We reviewed the recent literature reports published over last decade, using various electronic database searches such as PubMed, Embase, Medline, Web of Science, and Google Scholar. Clinical trial data till 2021 was retrieved from ClinicalTrial.gov. Major chemical classes developed as selective KSP inhibitors include dihydropyrimidines, β-carbolines, carbazoles, benzimidazoles, fused aryl derivatives, pyrimidines, fused pyrimidines, quinazolines, quinolones, thiadiazolines, spiropyran and azobenzenes. Drugs such as filanesib, litronesib, ispinesib have entered clinical trials, the most advanced phase explored being Phase II. KSP inhibitors have exhibited promising results; however, continued exploration is greatly required to establish the clinical potential of KSP inhibitors.
Collapse
Affiliation(s)
- Rinkal Chamariya
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.L. Mehta Road, Vile Parle (West), Mumbai - 400056, Maharashtra, India
| | - Vasanti Suvarna
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.L. Mehta Road, Vile Parle (West), Mumbai - 400056, Maharashtra, India
| |
Collapse
|
2
|
Gonçalves IL, Rockenbach L, das Neves GM, Göethel G, Nascimento F, Porto Kagami L, Figueiró F, Oliveira de Azambuja G, de Fraga Dias A, Amaro A, de Souza LM, da Rocha Pitta I, Avila DS, Kawano DF, Garcia SC, Battastini AMO, Eifler-Lima VL. Effect of N-1 arylation of monastrol on kinesin Eg5 inhibition in glioma cell lines. MEDCHEMCOMM 2018; 9:995-1010. [PMID: 30108989 PMCID: PMC6072436 DOI: 10.1039/c8md00095f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/14/2018] [Indexed: 01/08/2023]
Abstract
An original and focused library of two sets of dihydropyrimidin-2-thiones (DHPMs) substituted with N-1 aryl groups derived from monastrol was designed and synthesized in order to discover a more effective Eg5 ligand than the template. Based on molecular docking studies, four ligands were selected to perform pharmacological investigations against two glioma cell lines. The results led to the discovery of two original compounds, called 20h and 20e, with an anti-proliferative effects, achieving IC50 values of about half that of the IC50 of monastrol in both cell lines. As with monastrol, flow cytometry analyses showed that the 20e and 20h compounds induced cell cycle arrest in the G2/M phase, and immunocytochemistry essays revealed the formation of monopolar spindles due to Eg5 inhibition without any toxicity to Caenorhabditis elegans.
Collapse
Affiliation(s)
- Itamar Luís Gonçalves
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| | - Liliana Rockenbach
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| | - Gustavo Machado das Neves
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| | - Gabriela Göethel
- Laboratório de Toxicologia - LATOX , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Porto Alegre/RS , Brazil
| | - Fabiana Nascimento
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| | - Luciano Porto Kagami
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| | - Fabrício Figueiró
- Departamento de Bioquímica , ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre/RS , Brazil .
| | - Gabriel Oliveira de Azambuja
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| | - Amanda de Fraga Dias
- Departamento de Bioquímica , ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre/RS , Brazil .
| | - Andressa Amaro
- Departamento de Bioquímica , ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre/RS , Brazil .
| | - Lauro Mera de Souza
- Instituto de Pesquisa Pelé Pequeno Príncipe , Faculdades Pequeno Príncipe , Curitiba-PR , Brazil
| | - Ivan da Rocha Pitta
- Núcleo de Pesquisa em Inovação Terapêutica , Universidade Federal de Pernambuco , Recife/PE , Brazil
| | - Daiana Silva Avila
- Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCE) , Universidade Federal do Pampa-UNIPAMPA , Uruguaiana , RS , Brazil
| | - Daniel Fábio Kawano
- Faculdade de Ciências Farmacêuticas , Universidade Estadual de Campinas , Campinas-SP , Brazil
| | - Solange Cristina Garcia
- Laboratório de Toxicologia - LATOX , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Porto Alegre/RS , Brazil
| | | | - Vera Lucia Eifler-Lima
- Laboratório de Síntese Orgânica Medicinal/LaSOM , Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Avenida Ipiranga , 2752 , Porto Alegre/RS , Brazil .
| |
Collapse
|
3
|
Raab M, Sanhaji M, Matthess Y, Hörlin A, Lorenz I, Dötsch C, Habbe N, Waidmann O, Kurunci-Csacsko E, Firestein R, Becker S, Strebhardt K. PLK1 has tumor-suppressive potential in APC-truncated colon cancer cells. Nat Commun 2018; 9:1106. [PMID: 29549256 PMCID: PMC5856809 DOI: 10.1038/s41467-018-03494-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/19/2018] [Indexed: 12/13/2022] Open
Abstract
The spindle assembly checkpoint (SAC) acts as a molecular safeguard in ensuring faithful chromosome transmission during mitosis, which is regulated by a complex interplay between phosphatases and kinases including PLK1. Adenomatous polyposis coli (APC) germline mutations cause aneuploidy and are responsible for familial adenomatous polyposis (FAP). Here we study the role of PLK1 in colon cancer cells with chromosomal instability promoted by APC truncation (APC-ΔC). The expression of APC-ΔC in colon cells reduces the accumulation of mitotic cells upon PLK1 inhibition, accelerates mitotic exit and increases the survival of cells with enhanced chromosomal abnormalities. The inhibition of PLK1 in mitotic, APC-∆C-expressing cells reduces the kinetochore levels of Aurora B and hampers the recruitment of SAC component suggesting a compromised mitotic checkpoint. Furthermore, Plk1 inhibition (RNAi, pharmacological compounds) promotes the development of adenomatous polyps in two independent Apc Min/+ mouse models. High PLK1 expression increases the survival of colon cancer patients expressing a truncated APC significantly.
Collapse
Affiliation(s)
- Monika Raab
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany
| | - Mourad Sanhaji
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany
| | - Yves Matthess
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany
- German Cancer Consortium (DKTK)/ German Cancer Research Center, 69120, Heidelberg, Germany
| | - Albrecht Hörlin
- Institute of Pathology at the Department of Pathology, Goethe-University, 60590, Frankfurt, Germany
| | - Ioana Lorenz
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany
| | - Christina Dötsch
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany
| | - Nils Habbe
- Department of General and Visceral Surgery, Goethe-University, 60590, Frankfurt, Germany
| | - Oliver Waidmann
- Department of Gastroenterology and Hepatology, Goethe-University, 60590, Frankfurt, Germany
| | | | - Ron Firestein
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, AU 31681, Australia
- Department of Molecular Translational Medicine, Monash University, Clayton, VIC, 3800, Australia
| | - Sven Becker
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Gynecology, Goethe-University, 60590, Frankfurt, Germany.
- German Cancer Consortium (DKTK)/ German Cancer Research Center, 69120, Heidelberg, Germany.
| |
Collapse
|
4
|
Makala H, Ulaganathan V. Identification of novel scaffolds to inhibit human mitotic kinesin Eg5 targeting the second allosteric binding site using in silico methods. J Recept Signal Transduct Res 2017; 38:12-19. [PMID: 29041840 DOI: 10.1080/10799893.2017.1387922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Human mitotic kinesins are potential anticancer drug targets because of their essential role in mitotic cell division. The kinesin Eg5 (Kinesin-5, kif11) has gained much attention in this regard and has many inhibitors in different phases of clinical trials. All drug candidates considered for Eg5 so far binds to the binding site (Site 1) formed by the loop L5, helices α2 and α3 and are uncompetitive to ATP/ADP. Recently, it has been reported that Eg5 also has a second binding site (Site 2) formed by helices α4 and α6. In the current work, we have screened the compounds in the diversity set-III from National Cancer Institute (NCI) and Zinc database to identify potential inhibitors for Eg5 that specifically binds to the site 2. The compounds were ranked based on the glide extra precision docking scores and the top ranked compounds were found to have pyridazine scaffold. The top five compounds were further evaluated for other drug like properties. Stability of protein-ligand complexes were analyzed using molecular dynamic simulations. Our studies suggest that pyridazine analogs have good MDCK, permeability properties and high binding affinity to the human Eg5.
Collapse
Affiliation(s)
- Himesh Makala
- a Department of Biotechnology , School of Chemical and Biotechnology, SASTRA University , Thanjavur , India
| | | |
Collapse
|
5
|
Lu M, Zhu H, Wang X, Zhang D, Xiong L, Xu L, You Y. The prognostic role of Eg5 expression in laryngeal squamous cell carcinoma. Pathology 2016; 48:214-8. [DOI: 10.1016/j.pathol.2016.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/09/2015] [Accepted: 11/11/2015] [Indexed: 12/28/2022]
|
6
|
Longchamp A, Tao M, Bartelt A, Ding K, Lynch L, Hine C, Corpataux JM, Kristal BS, Mitchell JR, Ozaki CK. Surgical injury induces local and distant adipose tissue browning. Adipocyte 2016; 5:163-74. [PMID: 27386152 DOI: 10.1080/21623945.2015.1111971] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 10/12/2015] [Accepted: 10/16/2015] [Indexed: 12/20/2022] Open
Abstract
The adipose organ, which comprises brown, white and beige adipocytes, possesses remarkable plasticity in response to feeding and cold exposure. The development of beige adipocytes in white adipose tissue (WAT), a process called browning, represents a promising route to treat metabolic disorders. While surgical procedures constantly traumatize adipose tissue, its impact on adipocyte phenotype remains to be established. Herein, we studied the effect of trauma on adipocyte phenotype one day after sham, incision control, or surgical injury to the left inguinal adipose compartment. Caloric restriction was used to control for surgery-associated body temperature changes and weight loss. We characterized the trauma-induced cellular and molecular changes in subcutaneous, visceral, interscapular, and perivascular adipose tissue using histology, immunohistochemistry, gene expression, and flow cytometry analysis. After one day, surgical trauma stimulated adipose tissue browning at the site of injury and, importantly, in the contralateral inguinal depot. Browning was not present after incision only, and was largely independent of surgery-associated body temperature and weight loss. Adipose trauma rapidly recruited monocytes to the injured site and promoted alternatively activated macrophages. Conversely, PDGF receptor-positive beige progenitors were reduced. In this study, we identify adipose trauma as an unexpected driver of selected local and remote adipose tissue browning, holding important implications for the biologic response to surgical injury.
Collapse
Affiliation(s)
- Alban Longchamp
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Ming Tao
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Alexander Bartelt
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Kui Ding
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Jean-Marc Corpataux
- Department of Thoracic and Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Bruce S. Kristal
- Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - C. Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Martin SK, Kyprianou N. Exploitation of the Androgen Receptor to Overcome Taxane Resistance in Advanced Prostate Cancer. Adv Cancer Res 2015; 127:123-58. [PMID: 26093899 DOI: 10.1016/bs.acr.2015.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prostate cancer is a tumor addicted to androgen receptor (AR) signaling, even in its castration resistant state, and recently developed antiandrogen therapies including Abiraterone acetate and enzalutamide effectively target the androgen signaling axis, but there is ultimately recurrence to lethal disease. Development of advanced castration-resistant prostate cancer (CRPC) is a biological consequence of lack of an apoptotic response of prostate tumor cells to androgen ablation. Taxanes represent the major clinically relevant chemotherapy for the treatment of patients with metastatic CRPC; unfortunately, they do not deliver a cure but an extension of overall survival. First-generation taxane chemotherapies, Docetaxel (Taxotere), effectively target the cytoskeleton by stabilizing the interaction of β-tubulin subunits of microtubules preventing depolymerization, inducing G2M arrest and apoptosis. Shifting the current paradigm is a growing evidence to indicate that Docetaxel can effectively target the AR signaling axis by blocking its nuclear translocation and transcriptional activity in androgen-sensitive and castration-resistant prostate cancer cells, implicating a new mechanism of cross-resistance between microtubule-targeting chemotherapy and antiandrogen therapies. More recently, Cabazitaxel has emerged as a second-line taxane chemotherapy capable of conferring additional survival benefit to patients with CRPC previously treated with Docetaxel or in combination with antiandrogens. Similar to Docetaxel, Cabazitaxel induces apoptosis and G2M arrest; in contrast to Docetaxel, it sustains AR nuclear accumulation although it reduces the overall AR levels and FOXO1 expression. Cabazitaxel treatment also leads to downregulation of the microtubule-depolymerizing mitotic kinesins, MCAK, and HSET, preventing their ability to depolymerize microtubules and thus enhancing sensitivity to taxane treatment. The molecular mechanisms underlying taxane resistance involve mutational alterations in the tubulin subunits, microtubule dynamics, phenotyping programming of the epithelial-to-mesenchymal transition landscape, and the status of AR activity. This chapter discusses the mechanisms driving the therapeutic resistance of taxanes and antiandrogen therapies in CRPC, and the role of AR in potential interventions toward overcoming such resistance in patients with advanced metastatic disease.
Collapse
Affiliation(s)
- Sarah K Martin
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Natasha Kyprianou
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, USA; Department of Urology, University of Kentucky College of Medicine, Lexington, Kentucky, USA; Department of Pathology and Toxicology, University of Kentucky College of Medicine, Lexington, Kentucky, USA; Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA.
| |
Collapse
|
8
|
Label-free quantitative proteomic analysis reveals potential biomarkers and pathways in renal cell carcinoma. Tumour Biol 2014; 36:939-51. [PMID: 25315187 DOI: 10.1007/s13277-014-2694-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/30/2014] [Indexed: 01/22/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common malignancies in adults, and there is still no acknowledged biomarker for its diagnosis, prognosis, recurrence monitoring, and treatment stratification. Besides, little is known about the post-translational modification (PTM) of proteins in RCC. Here, we performed quantitative proteomic analysis on 12 matched pairs of clear cell RCC (ccRCC) and adjacent kidney tissues using liquid chromatography-tandem mass spectrometry (nanoLCMS/MS) and Progenesis LC-MS software (label-free) to identify and quantify the dysregulated proteins. A total of 1872 and 1927 proteins were identified in ccRCC and adjacent kidney tissues, respectively. Among these proteins, 1037 proteins were quantified by Progenesis LC-MS, and 213 proteins were identified as dysregulated proteins between ccRCC and adjacent tissues. Pathway analysis using IPA, STRING, and David tools was performed, which demonstrated the enrichment of cancer-related signaling pathways and biological processes such as mitochondrial dysfunction, metabolic pathway, cell death, and acetylation. Dysregulation of two mitochondrial proteins, acetyl-CoA acetyltransferase 1 (ACAT1) and manganese superoxide dismutase (MnSOD) were selected and confirmed by Western blotting and immunohistochemistry assays using another 6 pairs of ccRCC and adjacent tissues. Further mass spectrometry analysis indicated that both ACAT1 and MnSOD had characterized acetylation at lysine residues, which is the first time to identify acetylation of ACAT1 and MnSOD in ccRCC. Collectively, these data revealed a number of dysregulated proteins and signaling pathways by label-free quantitative proteomic approach in RCC, which shed light on potential diagnostic or prognostic biomarkers and therapeutic molecular targets for clinical intervention of RCC.
Collapse
|