1
|
Velavalapalli VM, Maddipati V, Gurská S, Annadurai N, Lišková B, Katari NK, Džubák P, Hajdúch M, Das V, Gundla R. Novel 5-Substituted Oxindole Derivatives as Bruton's Tyrosine Kinase Inhibitors: Design, Synthesis, Docking, Molecular Dynamics Simulation, and Biological Evaluation. ACS OMEGA 2024; 9:8067-8081. [PMID: 38405484 PMCID: PMC10882696 DOI: 10.1021/acsomega.3c08343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/07/2024] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
Bruton's tyrosine kinase (BTK) is a non-RTK cytoplasmic kinase predominantly expressed by hemopoietic lineages, particularly B-cells. A new oxindole-based focused library was designed to identify potent compounds targeting the BTK protein as anticancer agents. This study used rational approaches like structure-based pharmacophore modeling, docking, and ADME properties to select compounds. Molecular dynamics simulations carried out at 20 ns supported the stability of compound 9g within the binding pocket. All the compounds were synthesized and subjected to biological screening on two BTK-expressing cancer cell lines, RAMOS and K562; six non-BTK cancer cell lines, A549, HCT116 (parental and p53-/-), U2OS, JURKAT, and CCRF-CEM; and two non-malignant fibroblast lines, BJ and MRC-5. This study resulted in the identification of four new compounds, 9b, 9f, 9g, and 9h, possessing free binding energies of -10.8, -11.1, -11.3, and -10.8 kcal/mol, respectively, and displaying selective cytotoxicity against BTK-high RAMOS cells. Further analysis demonstrated the antiproliferative activity of 9h in RAMOS cells through selective inhibition of pBTK (Tyr223) without affecting Lyn and Syk, upstream proteins in the BCR signaling pathway. In conclusion, we identified a promising oxindole derivative (9h) that shows specificity in modulating BTK signaling pathways.
Collapse
Affiliation(s)
- Vani Madhuri Velavalapalli
- GITAM
School of Pharmacy, GITAM Deemed to Be University, Hyderabad, Telangana 502329, India
- Department
of Chemistry, GITAM School of Science, GITAM
Deemed to Be University, Hyderabad, Telangana 502329, India
| | | | - Soňa Gurská
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital
Olomouc, Hněvotínská
1333/5, Olomouc 77900, Czech Republic
- Czech
Advanced Technologies and Research Institute (CATRIN), Institute of
Molecular and Translational Medicine, Palacký
University Olomouc, Olomouc 77900, Czech Republic
| | - Narendran Annadurai
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital
Olomouc, Hněvotínská
1333/5, Olomouc 77900, Czech Republic
| | - Barbora Lišková
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital
Olomouc, Hněvotínská
1333/5, Olomouc 77900, Czech Republic
| | - Naresh Kumar Katari
- Department
of Chemistry, GITAM School of Science, GITAM
Deemed to Be University, Hyderabad, Telangana 502329, India
| | - Petr Džubák
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital
Olomouc, Hněvotínská
1333/5, Olomouc 77900, Czech Republic
- Czech
Advanced Technologies and Research Institute (CATRIN), Institute of
Molecular and Translational Medicine, Palacký
University Olomouc, Olomouc 77900, Czech Republic
| | - Marián Hajdúch
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital
Olomouc, Hněvotínská
1333/5, Olomouc 77900, Czech Republic
- Czech
Advanced Technologies and Research Institute (CATRIN), Institute of
Molecular and Translational Medicine, Palacký
University Olomouc, Olomouc 77900, Czech Republic
| | - Viswanath Das
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital
Olomouc, Hněvotínská
1333/5, Olomouc 77900, Czech Republic
- Czech
Advanced Technologies and Research Institute (CATRIN), Institute of
Molecular and Translational Medicine, Palacký
University Olomouc, Olomouc 77900, Czech Republic
| | - Rambabu Gundla
- Department
of Chemistry, GITAM School of Science, GITAM
Deemed to Be University, Hyderabad, Telangana 502329, India
| |
Collapse
|
2
|
Salerno S, Barresi E, Baglini E, Poggetti V, Da Settimo F, Taliani S. Target-Based Anticancer Indole Derivatives for the Development of Anti-Glioblastoma Agents. Molecules 2023; 28:molecules28062587. [PMID: 36985576 PMCID: PMC10056347 DOI: 10.3390/molecules28062587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor, with a poor prognosis and the highest mortality rate. Currently, GBM therapy consists of surgical resection of the tumor, radiotherapy, and adjuvant chemotherapy with temozolomide. Consistently, there are poor treatment options and only modest anticancer efficacy is achieved; therefore, there is still a need for the development of new effective therapies for GBM. Indole is considered one of the most privileged scaffolds in heterocyclic chemistry, so it may serve as an effective probe for the development of new drug candidates against challenging diseases, including GBM. This review analyzes the therapeutic benefit and clinical development of novel indole-based derivatives investigated as promising anti-GBM agents. The existing indole-based compounds which are in the pre-clinical and clinical stages of development against GBM are reported, with particular reference to the most recent advances between 2013 and 2022. The main mechanisms of action underlying their anti-GBM efficacy, such as protein kinase, tubulin and p53 pathway inhibition, are also discussed. The final goal is to pave the way for medicinal chemists in the future design and development of novel effective indole-based anti-GBM agents.
Collapse
|
3
|
Liao S, Yuk N, Kim YJ, Xu H, Li X, Wang L, Liu Y, Jung HJ. Novel terpestacin derivatives with l-amino acid residue as anticancer agents against U87MG-derived glioblastoma stem cells. Bioorg Chem 2023; 132:106392. [PMID: 36709667 DOI: 10.1016/j.bioorg.2023.106392] [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: 11/08/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
Based on the natural product terpestacin, seventeen derivatives (1-17) with various l-amino acid side chains were designed and synthesized. Their anticancer activities against U87MG-derived glioblastoma stem cells (GSCs) were evaluated, and compounds 5, 11, 13 and 15 showed strong abilities to inhibit the proliferation (IC50 = 2.8-6.9 μM) and tumorsphere formation of GSCs. Besides, compounds 13 and 15 could effectively induce apoptosis and significantly inhibit the invasion of GSCs (95 and 97 % inhibition, respectively, at 2.5 μM). The levels of CD133 marker in GSCs also decreased in dose-dependent manners after the treatment of these active compounds. Compared to terpestacin and the positive control A1938, our derivatives showed stronger activities and compounds 13 and 15 are promising candidates for further development as anticancer agents by targeting GSCs.
Collapse
Affiliation(s)
- Shengrong Liao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Research Center for Marine Microbes, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Nayeong Yuk
- Department of Pharmaceutical Engineering and Biotechnology, Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Korea
| | - Yu Jin Kim
- Department of Pharmaceutical Engineering and Biotechnology, Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Korea
| | - Huayan Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaolin Li
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Research Center for Marine Microbes, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Wang
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yonghong Liu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Research Center for Marine Microbes, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Hye Jin Jung
- Department of Pharmaceutical Engineering and Biotechnology, Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Korea.
| |
Collapse
|
4
|
Hsu JY, Hsu KC, Sun C, Chou CH, Lin TE, Sung TY, Tseng HJ, Yen SC, Yang CR, Huang WJ. Design, synthesis, and biological evaluation of indolin-2-one derivatives as novel cyclin-dependent protein kinase 8 (CDK8) inhibitors. Biomed Pharmacother 2023; 159:114258. [PMID: 36708700 DOI: 10.1016/j.biopha.2023.114258] [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: 11/22/2022] [Revised: 01/10/2023] [Accepted: 01/14/2023] [Indexed: 01/27/2023] Open
Abstract
Cyclin-dependent protein kinase 8 (CDK8) plays important roles in regulating fibrotic growth factors and inflammatory signaling pathways. Long-term chronic inflammation of the lungs can lead to idiopathic pulmonary fibrosis (IPF). Abnormal alveolar epithelial regeneration leads to the release of various fibrotic growth factors and the activation of inflammatory cells. CDK8 regulates profibrotic cytokines broadly implicated in the pathogenesis of fibrosis. Therefore, inhibition of CDK8 is considered a promising strategy for treating IPF. Here, CDK8 inhibitors were designed and optimized using a fragment-based drug design strategy. Testing results revealed that 71% of the synthesized compounds inhibited CDK8 activity better than the original compound E966-0530. Of these compounds, compound 4k exhibited the strongest CDK8 enzyme-inhibiting activity (IC50 =129 nM). Notably, it displayed a 13-fold increase in potency when compared to E966-0530. Experiments on toxicity and inhibition of epithelial-mesenchymal transition (EMT) protein expressions showed that compound 4k can inhibit EMT protein expressions, but with no significant cytotoxicity for alveolar epithelial cells. Compound 4k showed a potent inhibitory effect in cell migration assays. Furthermore, compound 4k significantly inhibited the phosphorylation of p-Smad3 and RNA Pol II, which are critical mediators in the fibrotic response signaling pathway. Compound 4k remarkably reduced TGF-β1-induced oxidative stress. The above results reveal optimized CDK8 inhibitors with potential use for IPF therapeutic treatment.
Collapse
Affiliation(s)
- Jui-Yi Hsu
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching Sun
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsuan Chou
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Ying Sung
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Hui-Ju Tseng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States
| | - Shih-Chung Yen
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong, People's Republic of China
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Wei-Jan Huang
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
5
|
Maddipati VC, Mittal L, Kaur J, Rawat Y, Koraboina CP, Bhattacharyya S, Asthana S, Gundla R. Discovery of non-nucleoside oxindole derivatives as potent inhibitors against dengue RNA-dependent RNA polymerase. Bioorg Chem 2023; 131:106277. [PMID: 36444792 DOI: 10.1016/j.bioorg.2022.106277] [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: 05/26/2022] [Revised: 10/20/2022] [Accepted: 11/12/2022] [Indexed: 11/21/2022]
Abstract
A series of thiazole linked Oxindole-5-Sulfonamide (OSA) derivatives were designed as inhibitors of RNA-dependent RNA polymerase (RdRp) activity of Dengue virus. These were synthesized and then evaluated for their efficacy in ex-vivo virus replication assay using human cell lines. Among 20 primary compounds in the series, OSA-15 was identified as a hit. A series of analogues were synthesized by replacing the difluoro benzyl group of OSA-15 with different substituted benzyl groups. The efficacy of OSA-15derivatives was less than that of the parent compound, except OSA-15-17, which has shown improved efficacy than OSA-15. The further optimization was carried out by adding dimethyl (DM) groups to both the sulfonamide and oxindole NH's to produce OSA-15-DM and OSA-15-17-DM. These two compounds were showing no detectable cytotoxicity and the latter was more efficacious. Further, both these compounds were tested for inhibition in all the serotypes of the Dengue virus using an ex-vivo assay. The EC50 of OSA-15-17-DM was observed in a low micromolar range between 2.5 and 5.0 µg/ml. Computation docking and molecular dynamics simulation studies confirmed the binding of identified hits to DENV RdRp. OSA15-17-DM blocks the RNA entrance and elongation site for their biological activity with high binding affinity. Overall, the identified oxindole derivatives are novel compounds that can inhibit Dengue replication, working as non-nucleoside inhibitors (NNI) to explore as anti-viral RdRp activity.
Collapse
Affiliation(s)
| | - Lovika Mittal
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3(rd)Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, Haryana, India
| | - Jaskaran Kaur
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3(rd)Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, Haryana, India
| | - Yogita Rawat
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3(rd)Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, Haryana, India
| | - Chandra Prakash Koraboina
- Department of Chemistry, School of Science, GITAM (Deemed to be University) Hyderabad, Telangana 502 329, India
| | - Sankar Bhattacharyya
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3(rd)Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, Haryana, India.
| | - Shailendra Asthana
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3(rd)Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, Haryana, India.
| | - Rambabu Gundla
- Department of Chemistry, School of Science, GITAM (Deemed to be University) Hyderabad, Telangana 502 329, India.
| |
Collapse
|
6
|
Thakur A, Faujdar C, Sharma R, Sharma S, Malik B, Nepali K, Liou JP. Glioblastoma: Current Status, Emerging Targets, and Recent Advances. J Med Chem 2022; 65:8596-8685. [PMID: 35786935 PMCID: PMC9297300 DOI: 10.1021/acs.jmedchem.1c01946] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Glioblastoma (GBM) is a highly malignant
brain tumor characterized
by a heterogeneous population of genetically unstable and highly infiltrative
cells that are resistant to chemotherapy. Although substantial efforts
have been invested in the field of anti-GBM drug discovery in the
past decade, success has primarily been confined to the preclinical
level, and clinical studies have often been hampered due to efficacy-,
selectivity-, or physicochemical property-related issues. Thus, expansion
of the list of molecular targets coupled with a pragmatic design of
new small-molecule inhibitors with central nervous system (CNS)-penetrating
ability is required to steer the wheels of anti-GBM drug discovery
endeavors. This Perspective presents various aspects of drug discovery
(challenges in GBM drug discovery and delivery, therapeutic targets,
and agents under clinical investigation). The comprehensively covered
sections include the recent medicinal chemistry campaigns embarked
upon to validate the potential of numerous enzymes/proteins/receptors
as therapeutic targets in GBM.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Chetna Faujdar
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida 201307, India
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Basant Malik
- Department of Sterile Product Development, Research and Development-Unit 2, Jubiliant Generics Ltd., Noida 201301, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| |
Collapse
|
7
|
Hassan R, Mohi-Ud-Din R, Dar MO, Shah AJ, Mir PA, Shaikh M, Pottoo FH. Bioactive Heterocyclic Compounds as Potential Therapeutics in the Treatment of Gliomas: A Review. Anticancer Agents Med Chem 2021; 22:551-565. [PMID: 34488596 DOI: 10.2174/1871520621666210901112954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most alarming diseases, with an estimation of 9.6 million deaths in 2018. Glioma occurs in glial cells surrounding nerve cells. The majority of the patients with gliomas have a terminal prognosis, and the ailment has significant sway on patients and their families, be it physical, psychological, or economic wellbeing. As Glioma exhibits, both intra and inter tumour heterogeneity with multidrug resistance and current therapies are ineffective. So the development of safer anti gliomas agents is the need of hour. Bioactive heterocyclic compounds, eithernatural or synthetic,are of potential interest since they have been active against different targets with a wide range of biological activities, including anticancer activities. In addition, they can cross the biological barriers and thus interfere with various signalling pathways to induce cancer cell death. All these advantages make bioactive natural compounds prospective candidates in the management of glioma. In this review, we assessed various bioactive heterocyclic compounds, such as jaceosidin, hispudlin, luteolin, silibinin, cannabidiol, tetrahydrocannabinol, didemnin B, thymoquinone, paclitaxel, doxorubicin, and cucurbitacins for their potential anti-glioma activity. Also, different kinds of chemical reactions to obtain various heterocyclic derivatives, e.g. indole, indazole, benzimidazole, benzoquinone, quinoline, quinazoline, pyrimidine, and triazine, are listed.
Collapse
Affiliation(s)
- Reyaz Hassan
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir. India
| | - Roohi Mohi-Ud-Din
- Pharmacognosy Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, Kashmir. India
| | - Mohammad Ovais Dar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Science and Research (NIPER), S.A.S. Nagar, Mohali, Punjab-160062. India
| | - Abdul Jalil Shah
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir. India
| | - Prince Ahad Mir
- Amritsar Pharmacy College, 12 KM stone Amritsar Jalandhar GT Road, Mandwala-143001. India
| | - Majeed Shaikh
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001. India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 31441, Dammam. Saudi Arabia
| |
Collapse
|
8
|
Sadeghi F, Afkhami A, Madrakian T, Ghavami R. Computational study on subfamilies of piperidine derivatives: QSAR modelling, model external verification, the inter-subset similarity determination, and structure-based drug designing. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2021; 32:433-462. [PMID: 33960256 DOI: 10.1080/1062936x.2021.1891568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/14/2021] [Indexed: 06/12/2023]
Abstract
A new subset of furan-pyrazole piperidine derivatives was used for QSAR model development. These compounds exhibit good Akt1 inhibitory activity; moreover, antiproliferative activities in vitro against OVCAR-8 (Human ovarian carcinoma cells) and HCT116 (human colon cancer cells), were confirmed for them. Based on the relevant three-dimensional (3D) and 2D autocorrelation descriptors, selected by genetic algorithm (GA), multiple linear regression (MLR) was established on half maximal-inhibitory concentration (IC50), in Akt1 and cancer cell lines independently. Robustness, stability, and predictive ability of the models were evaluated using external and internal validation (r2: 0.742-0.832, Q2LOO: 0.684-0.796, RMSE: 0.247-0.299, F: 32.283-57.578, and r2y-random: 0.049-0.080). Furthermore, in the new strategy, each of the evaluated models was generalized to two other subfamilies of piperidines to simultaneously compare the activities and structural similarity of these three subsets. Probably, structural similarity can be more considered as a criterion of similarity in the mechanism of action. Also, external verification of suggested predictive models was performed by another subset. Finally, by focusing on M64 as the most potent in vivo antitumor compound, 15 new derivatives were designed and six potent candidates were proposed for further investigation.
Collapse
Affiliation(s)
- F Sadeghi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran
| | - A Afkhami
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran
- Department of Chemistry, D-8 International University, Hamedan, Iran
| | - T Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran
| | - R Ghavami
- Chemometrics Laboratory, Chemistry Department, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| |
Collapse
|
9
|
Small Molecules of Marine Origin as Potential Anti-Glioma Agents. Molecules 2021; 26:molecules26092707. [PMID: 34063013 PMCID: PMC8124757 DOI: 10.3390/molecules26092707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022] Open
Abstract
Marine organisms are able to produce a plethora of small molecules with novel chemical structures and potent biological properties, being a fertile source for discovery of pharmacologically active compounds, already with several marine-derived agents approved as drugs. Glioma is classified by the WHO as the most common and aggressive form of tumor on CNS. Currently, Temozolomide is the only chemotherapeutic option approved by the FDA even though having some limitations. This review presents, for the first time, a comprehensive overview of marine compounds described as anti-glioma agents in the last decade. Nearly fifty compounds were compiled in this document and organized accordingly to their marine sources. Highlights on the mechanism of action and ADME properties were included. Some of these marine compounds could be promising leads for the discovery of new therapeutic alternatives for glioma treatment.
Collapse
|
10
|
Dhokne P, Sakla AP, Shankaraiah N. Structural insights of oxindole based kinase inhibitors as anticancer agents: Recent advances. Eur J Med Chem 2021; 216:113334. [PMID: 33721669 DOI: 10.1016/j.ejmech.2021.113334] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022]
Abstract
Small-molecule kinase inhibitors are being continuously explored as new anticancer therapeutics. Kinases are the phosphorylating enzymes which regulate numerous cellular functions such as proliferation, differentiation, migration, metabolism, and angiogenesis by activating several signalling pathways. Kinases have also been frequently found to be deregulated and overexpressed in cancerous tissues. Therefore, modulating the kinase activity by employing small molecules has emerged as a strategic approach for cancer treatment. On the other hand, oxindole motifs have surfaced as privileged scaffolds with significant multi-kinase inhibitory activity. The present review summarises recent advances in the development of oxindole based kinase inhibitors. The role of distinguished structural frameworks of oxindoles, such as 3-alkenyl oxindoles, spirooxindoles, 3-iminooxindoles and similar hydrazone derivatives have been described based on their kinase inhibition potential. Furthermore, the design strategies, mechanism of actions, structure activity relationships (SARs) and their mode of interaction with target protein have been critically highlighted.
Collapse
Affiliation(s)
- Prajwal Dhokne
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Akash P Sakla
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India.
| |
Collapse
|
11
|
Liu M, Ju X, Zou J, Shi J, Jia G. Recent researches for dual Aurora target inhibitors in antitumor field. Eur J Med Chem 2020; 203:112498. [PMID: 32693295 DOI: 10.1016/j.ejmech.2020.112498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/28/2020] [Indexed: 11/17/2022]
Abstract
Non-infectious and chronic diseases such as malignant tumors are now one of the main causes of human death. Its occurrence is a multi-factor, multi-step complex process with biological characteristics such as cell differentiation, abnormal proliferation, uncontrolled growth, and metastasis. It has been found that a variety of human malignant tumors are accompanied by over-expression and proliferation of Aurora kinase, which causes abnormalities in the mitotic process and is related to the instability of the genome that causes tumors. Therefore, the use of Aurora kinase inhibitors to target tumors is becoming a research hotspot. However, in cancer, because of the complexity of signal transduction system and the participation of different proteins and enzymes, the anticancer effect of selective single-target drugs is limited. After inhibiting one pathway, signal molecules can be conducted through other pathways, resulting in poor therapeutic effect of single-target drug treatment. Multi-target drugs can solve this problem very well. It can regulate the various links that cause disease at the same time without completely eliminating the relationship between the signal transmission systems, and it is not easy to cause drug resistance. Currently, studies have shown that Aurora dual-target inhibitors generated with the co-inhibition of Aurora and another target (such as CDK, PLK, JAK2, etc.) have better therapeutic effects on tumors. In this paper, we reviewed the studies of dual Aurora inhibitors that have been discovered in recent years.
Collapse
Affiliation(s)
- Maoyu Liu
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xueming Ju
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jing Zou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
12
|
Chaudhari MB, Jayan K, Gnanaprakasam B. Sn-Catalyzed Criegee-Type Rearrangement of Peroxyoxindoles Enabled by Catalytic Dual Activation of Esters and Peroxides. J Org Chem 2020; 85:3374-3382. [DOI: 10.1021/acs.joc.9b03160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Moreshwar B. Chaudhari
- Department of Chemistry, Indian Institute of Science Education and Research, Pune 411008, India
| | - Krishna Jayan
- Department of Chemistry, Indian Institute of Science Education and Research, Pune 411008, India
| | - Boopathy Gnanaprakasam
- Department of Chemistry, Indian Institute of Science Education and Research, Pune 411008, India
| |
Collapse
|
13
|
Preparation and biological evaluation of quinoline amines as anticancer agents and its molecular docking. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02374-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
14
|
Fernandes GFDS, Fernandes BC, Valente V, Dos Santos JL. Recent advances in the discovery of small molecules targeting glioblastoma. Eur J Med Chem 2018; 164:8-26. [PMID: 30583248 DOI: 10.1016/j.ejmech.2018.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is one of the most common central nervous system cancers. It is characterized as a fast-growing tumor that arises from multiple cell types with neural stem-cell-like properties. Additionally, GBM tumors are highly invasive, which is attributed to the presence of glioblastoma stem cells that makes surgery ineffective in most cases. Currently, temozolomide is the unique chemotherapy option approved by the U.S. Food and Drug Administration for GBM treatment. This review analyzes the emergence and development of new synthetic small molecules discovered as promising anti-glioblastoma agents. A number of compounds were described herein and grouped according to the main chemical class used in the drug discovery process. Importantly, we focused only on synthetic compounds published in the last 10 years, thus excluding natural products. Furthermore, we included in this review only those most biologically active compounds with proven in vitro and/or in vivo efficacy.
Collapse
Affiliation(s)
- Guilherme Felipe Dos Santos Fernandes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, 14800-060, Brazil
| | - Barbara Colatto Fernandes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Valeria Valente
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, 14800-060, Brazil.
| |
Collapse
|
15
|
Vennila K, Sunny D, Madhuri S, Ciattini S, Chelazzi L, Elango KP. Design, synthesis, crystal structures and anticancer activity of 4-substituted quinolines to target PDK1. Bioorg Chem 2018; 81:184-190. [DOI: 10.1016/j.bioorg.2018.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
|
16
|
Güçlü D, Kuzu B, Tozlu İ, Taşpınar F, Canpınar H, Taşpınar M, Menges N. Synthesis of novel imidazopyridines and their biological evaluation as potent anticancer agents: A promising candidate for glioblastoma. Bioorg Med Chem Lett 2018; 28:2647-2651. [PMID: 30042044 DOI: 10.1016/j.bmcl.2018.06.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/13/2018] [Accepted: 06/16/2018] [Indexed: 11/25/2022]
Abstract
Novel imidazopyridine derivatives were synthesized according to a very simple protocol and then subjected to cytotoxicity testing against LN-405 cells. Two of the compounds exhibited antiproliferative effects on LN-405 cells at 10 and 75 µM and were selected as lead compounds for further study. Safety experiment for lead compounds on WS1 was carried out and IC50 values were calculated as 480 and 844 µM. LN-405 cell line were incubated with the lead compounds and then tested for DNA damage by comet assay and effects on cell cycle using flow cytometry. The results of these two tests showed that both lead compounds affected the G0/G1 phase and did not allow the cells to reach the synthesis phase. The log BB (blood-brain barrier) and Caco-2 permeability of the synthesized molecules were calculated and it was shown that imidazopyridine derivatives taken orally are likely to pass through gastrointestinal membrane and the blood-brain barrier.
Collapse
Affiliation(s)
- Dilek Güçlü
- Faculty of Education, Chemistry Section, Van Yuzuncu Yil University, 65080 Van, Turkey; Pharmaceutical Chemistry Section, Faculty of Pharmacy, Van Yuzuncu Yıl University, 65080 Van, Turkey
| | - Burak Kuzu
- Pharmaceutical Chemistry Section, Faculty of Pharmacy, Van Yuzuncu Yıl University, 65080 Van, Turkey; SAFF Chemical Reagents R&D Lab., YYU-TEKNOKENT, 65080 Van, Turkey
| | - İsrafil Tozlu
- Faculty of Education, Chemistry Section, Van Yuzuncu Yil University, 65080 Van, Turkey
| | - Filiz Taşpınar
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, 65080 Van, Turkey
| | - Hande Canpınar
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Mehmet Taşpınar
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, 65080 Van, Turkey
| | - Nurettin Menges
- Pharmaceutical Chemistry Section, Faculty of Pharmacy, Van Yuzuncu Yıl University, 65080 Van, Turkey; SAFF Chemical Reagents R&D Lab., YYU-TEKNOKENT, 65080 Van, Turkey.
| |
Collapse
|
17
|
Daniele S, Pietrobono D, Costa B, Giustiniano M, La Pietra V, Giacomelli C, La Regina G, Silvestri R, Taliani S, Trincavelli ML, Da Settimo F, Novellino E, Martini C, Marinelli L. Bax Activation Blocks Self-Renewal and Induces Apoptosis of Human Glioblastoma Stem Cells. ACS Chem Neurosci 2018; 9:85-99. [PMID: 28368610 DOI: 10.1021/acschemneuro.7b00023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is characterized by a poor response to conventional chemotherapeutic agents, attributed to the insurgence of drug resistance mechanisms and to the presence of a subpopulation of glioma stem cells (GSCs). GBM cells and GSCs present, among others, an overexpression of antiapoptotic proteins and an inhibition of pro-apoptotic ones, which help to escape apoptosis. Among pro-apoptotic inducers, the Bcl-2 family protein Bax has recently emerged as a promising new target in cancer therapy along with first BAX activators (BAM7, Compound 106, and SMBA1). Herein, a derivative of BAM-7, named BTC-8, was employed to explore the effects of Bax activation in different human GBM cells and in their stem cell subpopulation. BTC-8 inhibited GBM cell proliferation, arrested the cell cycle, and induced apoptosis through the induction of mitochondrial membrane permeabilization. Most importantly, BTC-8 blocked proliferation and self-renewal of GSCs and induced their apoptosis. Notably, BTC-8 was demonstrated to sensitize both GBM cells and GSCs to the alkylating agent Temozolomide. Overall, our findings shed light on the effects and the relative molecular mechanisms related to Bax activation in GBM, and they suggest Bax-targeting compounds as promising therapeutic tools against the GSC reservoir.
Collapse
Affiliation(s)
- Simona Daniele
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Barbara Costa
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Valeria La Pietra
- Department
of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy
| | | | - Giuseppe La Regina
- Istituto
Pasteur Italia—Fondazione Cenci Bolognetti, Dipartimento di
Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, 00185 Roma, Italy
| | - Romano Silvestri
- Istituto
Pasteur Italia—Fondazione Cenci Bolognetti, Dipartimento di
Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, 00185 Roma, Italy
| | - Sabrina Taliani
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | | | - Ettore Novellino
- Department
of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy
| | - Claudia Martini
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Luciana Marinelli
- Department
of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy
| |
Collapse
|
18
|
Zaryanova EV, Ignatov AA, Lozynskaya NA. Synthesis and reactivity of new amide-substituted oxindole derivatives. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.10.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Targeting PDK1 for Chemosensitization of Cancer Cells. Cancers (Basel) 2017; 9:cancers9100140. [PMID: 29064423 PMCID: PMC5664079 DOI: 10.3390/cancers9100140] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023] Open
Abstract
Despite the rapid development in the field of oncology, cancer remains the second cause of mortality worldwide, with the number of new cases expected to more than double in the coming years. Chemotherapy is widely used to decelerate or stop tumour development in combination with surgery or radiation therapy when appropriate, and in many cases this improves the symptomatology of the disease. Unfortunately though, chemotherapy is not applicable to all patients and even when it is, there are many cases where a successful initial treatment period is followed by chemotherapeutic drug resistance. This is caused by a number of reasons, ranging from the genetic background of the patient (innate resistance) to the formation of tumour-initiating cells (acquired resistance). In this review, we discuss the potential role of PDK1 in the development of chemoresistance in different types of malignancy, and the design and application of potent inhibitors which can promote chemosensitization.
Collapse
|
20
|
Yang TH, Lee CI, Huang WH, Lee AR. Structural optimization and evaluation of novel 2-pyrrolidone-fused (2-oxoindolin-3-ylidene)methylpyrrole derivatives as potential VEGFR-2/PDGFRβ inhibitors. Chem Cent J 2017; 11:72. [PMID: 29086859 PMCID: PMC5539068 DOI: 10.1186/s13065-017-0301-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/20/2017] [Indexed: 12/21/2022] Open
Abstract
Background Tumor angiogenesis, essential for tumor growth and metastasis, is tightly regulated by VEGF/VEGFR and PDGF/PDGFR pathways, and therefore blocking those pathways is a promising therapeutic target. Compared to sunitinib, the C(5)-Br derivative of 2-pyrrolidone-fused (2-oxoindolin-3-ylidene)methylpyrrole has significantly greater in vitro activities against VEGFR-2, PDGFRβ, and tube formation. Results and discussion The objective of this study was to perform further structural optimization, which revealed certain new products with even more potent anti-tumor activities, both cellularly and enzymatically. Of these, 15 revealed ten- and eightfold stronger potencies against VEGFR-2 and PDGFRβ than sunitinib, respectively, and showed selectivity against HCT116 with a favorable selective index (SI > 4.27). The molecular docking results displayed that the ligand–protein binding affinity to VEGFR-2 could be enhanced by introducing a hydrogen-bond-donating (HBD) substituent at C(5) of (2-oxoindolin-3-ylidene)methylpyrrole such as 14 (C(5)-OH) and 15 (C(5)-SH). Conclusions Among newly synthetic compounds, 7 and 13–15 exhibited significant inhibitory activities against VEGFR-2 and PDGFRβ. Of these, the experimental results suggest that 15 might be a promising anti-proliferative agent. IC50 comparison of sunitinib, 14, and 15 against VEGFR-2 and PDGFRβ. ![]() Electronic supplementary material The online version of this article (doi:10.1186/s13065-017-0301-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting-Hsuan Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, No. 161, Section 6, Mingchuan East Road, Taipei, 11490, Taiwan
| | - Chun-I Lee
- School of Pharmacy, National Defense Medical Center, No. 161, Section 6, Mingchuan East Road, Taipei, 11490, Taiwan
| | - Wen-Hsin Huang
- School of Pharmacy, National Defense Medical Center, No. 161, Section 6, Mingchuan East Road, Taipei, 11490, Taiwan
| | - An-Rong Lee
- Graduate Institute of Medical Sciences, National Defense Medical Center, No. 161, Section 6, Mingchuan East Road, Taipei, 11490, Taiwan. .,School of Pharmacy, National Defense Medical Center, No. 161, Section 6, Mingchuan East Road, Taipei, 11490, Taiwan.
| |
Collapse
|
21
|
Gao Y, Ye DY, Zhou WC, Chu Y. The discovery of novel benzothiazinones as highly selective non-ATP competitive glycogen synthase kinase 3β inhibitors for the treatment of ovarian cancer. Eur J Med Chem 2017; 135:370-381. [DOI: 10.1016/j.ejmech.2017.04.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 01/08/2023]
|
22
|
Design, synthesis and biological evaluation of 3-substituted-2-oxindole hybrid derivatives as novel anticancer agents. Eur J Med Chem 2017; 134:258-270. [PMID: 28419928 DOI: 10.1016/j.ejmech.2017.03.089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/24/2017] [Accepted: 03/25/2017] [Indexed: 01/01/2023]
Abstract
The 2-oxindole nucleus is the central core to develop new anticancer agents and its substitution at the 3-position can effect antitumor activity. Utilizing a pharmacophore hybridization approach, a novel series of antiproliferative agents was obtained by the modification of the structure of 3-substituted-2-oxindole pharmacophore by the attachment of the α-bromoacryloyl moiety, acting as a Michael acceptor, at the 5-position of 2-oxindole framework. The impact of the substituent at the 3-position of 2-oxindole core on the potency and selectivity against a panel of seven different cancer cell lines was examined. We found that these hybrid molecules displayed potent antiproliferative activity against a panel of four cancer cell lines, with one-to double digit nanomolar 50% inhibitory concentrations (IC50). A distinctive selective antiproliferative activity was obtained towards CCRF-CEM and RS4; 11 leukemic cell lines. In order to study the possible mechanism of action, we observed that the two most active compounds namely 3(E) and 6(Z) strongly induce apoptosis that follow the mitochondrial pathway. Interestingly a decrease of intracellular reduced glutathione content (GSH) and reactive oxygen species (ROS) production was detected in treated cells compared with controls suggesting that these effects may be involved in their mechanism of action.
Collapse
|
23
|
Yue P, Gao L, Wang X, Ding X, Teng J. Intranasal Administration of GDNF Protects Against Neural Apoptosis in a Rat Model of Parkinson’s Disease Through PI3K/Akt/GSK3β Pathway. Neurochem Res 2017; 42:1366-1374. [DOI: 10.1007/s11064-017-2184-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 12/16/2022]
|
24
|
Daniele S, Sestito S, Pietrobono D, Giacomelli C, Chiellini G, Di Maio D, Marinelli L, Novellino E, Martini C, Rapposelli S. Dual Inhibition of PDK1 and Aurora Kinase A: An Effective Strategy to Induce Differentiation and Apoptosis of Human Glioblastoma Multiforme Stem Cells. ACS Chem Neurosci 2017; 8:100-114. [PMID: 27797168 DOI: 10.1021/acschemneuro.6b00251] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of glioblastoma multiforme (GBM) is mainly attributed to drug resistance mechanisms and to the existence of a subpopulation of glioma stem cells (GSCs). Multitarget compounds able to both affect different deregulated pathways and the GSC subpopulation could escape tumor resistance and, most importantly, eradicate the stem cell reservoir. In this respect, the simultaneous inhibition of phosphoinositide-dependent kinase-1 (PDK1) and aurora kinase A (AurA), each one playing a pivotal role in cellular survival/migration/differentiation, could represent an innovative strategy to overcome GBM resistance and recurrence. Herein, the cross-talk between these pathways was investigated, using the single-target reference compounds MP7 (PDK1 inhibitor) and Alisertib (AurA inhibitor). Furthermore, a new ligand, SA16, was identified for its ability to inhibit the PDK1 and the AurA pathways at once, thus proving to be a useful tool for the simultaneous inhibition of the two kinases. SA16 blocked GBM cell proliferation, reduced tumor invasiveness, and triggered cellular apoptosis. Most importantly, the AurA/PDK1 blocker showed an increased efficacy against GSCs, inducing their differentiation and apoptosis. To the best of our knowledge, this is the first report on combined targeting of PDK1 and AurA. This drug represents an attractive multitarget lead scaffold for the development of new potential treatments for GBM and GSCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Danilo Di Maio
- Scuola Normale Superiore, Piazza
dei Cavalieri 7, I-56126 Pisa, Italy
| | - Luciana Marinelli
- Department
of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Ettore Novellino
- Department
of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | | |
Collapse
|
25
|
Abstract
Glioblastoma (GBM) is the most aggressive of primary brain tumors. Despite the progress in understanding the biology of the pathogenesis of glioma made during the past decade, the clinical outcome of patients with GBM remains still poor. Deregulation of many signaling pathways involved in growth, survival, migration and resistance to treatment has been implicated in pathogenesis of GBM. One of these pathways is phosphatidylinositol-3 kinases (PI3K)/protein kinase B (AKT)/rapamycin-sensitive mTOR-complex (mTOR) pathway, intensively studied and widely described so far. Much less attention has been paid to the role of glycogen synthase kinase 3 β (GSK3β), a target of AKT. In this review we focus on the function of AKT/GSK3β signaling in GBM.
Collapse
|
26
|
Zhan W, Xu L, Dong X, Dong J, Yi X, Ma X, Qiu N, Li J, Yang B, Zhou Y, Hu Y. Design, synthesis and biological evaluation of pyrazol-furan carboxamide analogues as novel Akt kinase inhibitors. Eur J Med Chem 2016; 117:47-58. [DOI: 10.1016/j.ejmech.2016.03.074] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/12/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
|
27
|
Arfeen M, Bhagat S, Patel R, Prasad S, Roy I, Chakraborti AK, Bharatam PV. Design, synthesis and biological evaluation of 5-benzylidene-2-iminothiazolidin-4-ones as selective GSK-3β inhibitors. Eur J Med Chem 2016; 121:727-736. [PMID: 27423119 DOI: 10.1016/j.ejmech.2016.04.075] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/26/2016] [Accepted: 04/28/2016] [Indexed: 01/16/2023]
Abstract
In this work, iminothiazolidin-4-one derivatives were explored as selective GSK-3β inhibitors. Molecular docking analysis was carried to design a series of compounds, which were synthesized using substituted thiourea, 2-bromoacetophenones and benzaldehydes. Out of the twenty five compounds synthesized during this work, the in vitro evaluation against GSK-3 led to the identification of nine compounds with activity in lower nano-molar range (2-85 nM). Further, in vitro evaluation against CDK-2 showed five compounds to be selective towards GSK-3.
Collapse
Affiliation(s)
- Minhajul Arfeen
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India
| | - Shweta Bhagat
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India
| | - Rahul Patel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India
| | - Shivcharan Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India
| | - Asit K Chakraborti
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India
| | - Prasad V Bharatam
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160062, Punjab, India.
| |
Collapse
|
28
|
Sestito S, Daniele S, Nesi G, Zappelli E, Di Maio D, Marinelli L, Digiacomo M, Lapucci A, Martini C, Novellino E, Rapposelli S. Locking PDK1 in DFG-out conformation through 2-oxo-indole containing molecules: Another tools to fight glioblastoma. Eur J Med Chem 2016; 118:47-63. [PMID: 27123901 DOI: 10.1016/j.ejmech.2016.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/01/2016] [Accepted: 04/02/2016] [Indexed: 01/05/2023]
Abstract
The phosphoinositide-dependent kinase-1 (PDK1) is one of the main components of the PI3K/Akt pathway. Also named the "master kinase" of the AGC family, PDK1 plays a critical role in tumorigenesis, by enhancing cell proliferation and inhibiting apoptosis, as well as in cell invasion and metastasis formation. Although there have been done huge efforts in discovering specific compounds targeting PDK1, nowadays no PDK1 inhibitor has yet entered the clinic. With the aim to pick out novel and potent PDK1 inhibitors, herein we report the design and synthesis of a new class of molecules obtained by merging the 2-oxo-indole nucleus with the 2-oxo-pyridonyl fragment, two moieties with high affinity for the PDK1 hinge region and its DFG-out binding site, respectively. To this purpose, a small series of compounds were synthesised and a tandem application of docking and Molecular Dynamic (MD) was employed to get insight into their mode of binding. The OXID-pyridonyl hybrid 8, possessing the lower IC50 (IC50 = 112 nM), was also tested against recombinant kinases involved in the PI3K/PDK1/Akt pathway and was subjected to vitro studies to evaluate the cytotoxicity and the inhibition of tumour cell migration. All together the results let us to consider 8, as a lead compound of a new generation of PDK1 inhibitors and encourage us to further studies in this direction.
Collapse
Affiliation(s)
- Simona Sestito
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Giulia Nesi
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Elisa Zappelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Danilo Di Maio
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy; Istituto Nazionale di Fisica Nucleare (INFN), Largo Bruno Pontecorvo 3, 56127 Pisa, Italy
| | | | - Maria Digiacomo
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Annalina Lapucci
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | | | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy.
| |
Collapse
|
29
|
Ellert-Miklaszewska A, Dallavalle S, Musso L, Martinet N, Wojnicki K, Kaminska B. Identification of new scaffolds with anti-tumor action toward human glioblastoma cells. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00477f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Compounds containing an isothiazolonaphthoquinone core and HDAC inhibitors with an indolyl-substituted biphenyl-4-yl-acrylohydroxamic acid are promising drug candidates against malignant brain tumors, glioblastomas.
Collapse
Affiliation(s)
- Aleksandra Ellert-Miklaszewska
- Laboratory of Molecular Biology
- Neurobiology Center
- The Nencki Institute of Experimental Biology of Polish Academy of Sciences
- 02-093 Warsaw
- Poland
| | - Sabrina Dallavalle
- Department of Food
- Environmental and Nutritional Sciences
- Division of Chemistry and Molecular Biology
- 20133 Milan
- Italy
| | - Loana Musso
- Department of Food
- Environmental and Nutritional Sciences
- Division of Chemistry and Molecular Biology
- 20133 Milan
- Italy
| | - Nadine Martinet
- CNRS UMR 7272
- Institut de Chimie
- Université de Nice-Sophia Antipolis
- Nice
- France
| | - Kamil Wojnicki
- Laboratory of Molecular Biology
- Neurobiology Center
- The Nencki Institute of Experimental Biology of Polish Academy of Sciences
- 02-093 Warsaw
- Poland
| | - Bozena Kaminska
- Laboratory of Molecular Biology
- Neurobiology Center
- The Nencki Institute of Experimental Biology of Polish Academy of Sciences
- 02-093 Warsaw
- Poland
| |
Collapse
|
30
|
Nagarsenkar A, Prajapti SK, Guggilapu SD, Birineni S, Sravanti Kotapalli S, Ummanni R, Babu BN. Investigation of triazole-linked indole and oxindole glycoconjugates as potential anticancer agents: novel Akt/PKB signaling pathway inhibitors. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00513b] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel triazole-linked indole and oxindole glycoconjugates as inhibitors of the Akt/PKB signaling pathway.
Collapse
Affiliation(s)
- Atulya Nagarsenkar
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Hyderabad
- India
| | - Santosh Kumar Prajapti
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Hyderabad
- India
| | - Sravanthi Devi Guggilapu
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Hyderabad
- India
| | - Swetha Birineni
- Center for Chemical Biology
- Indian Institute of Chemical Technology (CSIR-IICT)
- Hyderabad 500007
- India
| | - Sudha Sravanti Kotapalli
- Center for Chemical Biology
- Indian Institute of Chemical Technology (CSIR-IICT)
- Hyderabad 500007
- India
| | - Ramesh Ummanni
- Center for Chemical Biology
- Indian Institute of Chemical Technology (CSIR-IICT)
- Hyderabad 500007
- India
| | - Bathini Nagendra Babu
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Hyderabad
- India
| |
Collapse
|