1
|
Choudhury R, Bahadi CK, Ray IP, Dash P, Pattanaik I, Mishra S, Mohapatra SR, Patnaik S, Nikhil K. PIM1 kinase and its diverse substrate in solid tumors. Cell Commun Signal 2024; 22:529. [PMID: 39487435 PMCID: PMC11531143 DOI: 10.1186/s12964-024-01898-y] [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: 08/19/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024] Open
Abstract
The PIM kinase family, consisting of PIM1, PIM2, and PIM3, is a group of serine/threonine protein kinases crucial for cellular growth, immunoregulation, and oncogenesis. PIM1 kinase is often overexpressed in solid and hematopoietic malignancies, promoting cell survival, proliferation, migration, and senescence by activating key genes. In vitro and in vivo studies have established the oncogenic potential of PIM1 kinases. These kinases have been implicated in tumor progression, metastasis, and resistance to chemotherapy, underscoring their potential as a therapeutic target for cancer therapy. This review delves into the intricate molecular mechanisms through which PIM1 interacts with specific substrates in different tumor tissues, leading to diverse outcomes in various human cancers. Over the past decade, the inhibition of PIM1 in cancers has garnered significant attention as a potential standalone treatment. Various in vitro, in vivo, and early clinical trial data have provided support for this approach to varying extents. Novel compounds that inhibit PIM1 kinase have shown effectiveness and a favorable toxicity profile in preclinical studies. Several of these substances are now being studied in clinical trials due to their promising outcomes. This article provides a thorough examination of the PIM1 kinase pathways and the recent advancements in producing PIM1 kinase inhibitors for the treatment of cancer.
Collapse
Affiliation(s)
- Rituparna Choudhury
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Chandan Kumar Bahadi
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Ipsa Pratibimbita Ray
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Pragyanshree Dash
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Isha Pattanaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Suman Mishra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Soumya R Mohapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Srinivas Patnaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Kumar Nikhil
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India.
| |
Collapse
|
2
|
Montero V, Montana M, Carré M, Vanelle P. Quinoxaline derivatives: Recent discoveries and development strategies towards anticancer agents. Eur J Med Chem 2024; 271:116360. [PMID: 38614060 DOI: 10.1016/j.ejmech.2024.116360] [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: 12/20/2023] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
Cancer is a leading cause of death and a major health problem worldwide. While many effective anticancer agents are available, most drugs currently on the market are not specific, raising issues like the common side effects of chemotherapy. However, recent research hold promises for the development of more efficient and safer anticancer drugs. Quinoxaline and its derivatives are becoming recognized as a novel class of chemotherapeutic agents with activity against different tumors. The present review compiles and discusses studies concerning the therapeutic potential of the anticancer activity of quinoxaline derivatives, covering articles published between January 2018 and January 2023.
Collapse
Affiliation(s)
- Vincent Montero
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, CEDEX 05, 13385, Marseille, France; AP-HM, Service de Pharmacologie Clinique et Pharmacovigilance, Hôpital de la Timone, Marseille CEDEX 05, 13385, France.
| | - Marc Montana
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, CEDEX 05, 13385, Marseille, France; AP-HM, Oncopharma, Hôpital Nord, Marseille, France
| | - Manon Carré
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm UMR1068, CNRS UMR7258, Aix-Marseille Université UM105, Institut Paoli Calmettes - Faculté de Pharmacie, Marseille, France
| | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, CEDEX 05, 13385, Marseille, France; AP-HM, Service Central de la Qualité et de l'Information Pharmaceutiques, Hôpital Conception, Marseille, 13005, France
| |
Collapse
|
3
|
Sharma A, Dubey R, Asati V, Baweja GS, Gupta S, Asati V. Assessment of structural and activity-related contributions of various PIM-1 kinase inhibitors in the treatment of leukemia and prostate cancer. Mol Divers 2024:10.1007/s11030-023-10795-4. [PMID: 38642309 DOI: 10.1007/s11030-023-10795-4] [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: 10/18/2023] [Accepted: 12/07/2023] [Indexed: 04/22/2024]
Abstract
One of the most perilous illnesses in the world is cancer. The cancer may be associated with the mutation of different genes inside the body. The PIM kinase, also known as the serine/threonine kinase, plays a critical role in the biology of different kinds of cancer. They are widely distributed and associated with several biological processes, including cell division, proliferation, and death. Aberration of PIM-1 kinase is found in varieties of cancer. Prostate cancer and leukemia can both be effectively treated with PIM-1 kinase inhibitors. There are several potent compounds that have been explored in this review based on heterocyclic compounds for the treatment of prostate cancer and leukemia that have strong effects on the suppression of PIM-1 kinase. The present review summarizes the PIM-1 kinase pathway, their inhibitors under clinical trial, related patents, and SAR studies of several monocyclic, bicyclic, and polycyclic compounds. The study related to their molecular interactions with receptors is also included in the present manuscript. The study may be beneficial to scientists for the development of novel compounds as PIM-1 inhibitors in the treatment of prostate cancer and leukemia.
Collapse
Affiliation(s)
- Anushka Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Rahul Dubey
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Vikas Asati
- Department of Medical Oncology, Sri Aurobindo Medical College and PG Institute, Indore, MP, India
| | - Gurkaran Singh Baweja
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Shankar Gupta
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
4
|
Chen L, Mao W, Ren C, Li J, Zhang J. Comprehensive Insights that Targeting PIM for Cancer Therapy: Prospects and Obstacles. J Med Chem 2024; 67:38-64. [PMID: 38164076 DOI: 10.1021/acs.jmedchem.3c01802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Proviral integration sitea for Moloney-murine leukemia virus (PIM) kinases are a family of highly conserved serine/tyrosine kinases consisting of three members, PIM-1, PIM-2, and PIM-3. These kinases regulate a wide range of substrates through phosphorylation and affect key cellular processes such as transcription, translation, proliferation, apoptosis, and energy metabolism. Several PIM inhibitors are currently undergoing clinical trials, such as a phase I clinical trial of Uzanserti (5) for the treatment of relapsed diffuse large B-cell lymphoma that has been completed. The current focus encompasses the structural and biological characterization of PIM, ongoing research progress on small-molecule inhibitors undergoing clinical trials, and evaluation analysis of persisting challenges in this field. Additionally, the design and discovery of small-molecule inhibitors targeting PIM in recent years have been explored, with a particular emphasis on medicinal chemistry, aiming to provide valuable insights for the future development of PIM inhibitors.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurology, Joint Research Institution of Altitude Health and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- 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, Sichuan, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Wuyu Mao
- Department of Neurology, Joint Research Institution of Altitude Health and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu 611130, Sichuan, China
| | - Jinqi Li
- 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, Sichuan, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
5
|
Yousra S, El Ghayati L, Hökelek T, Ouazzani Chahdi F, Mague JT, Kandri Rodi Y, Sebbar NK. Crystal structure and Hirshfeld surface analysis of 3-eth-oxy-1-ethyl-6-nitro-quinoxalin-2(1 H)-one. Acta Crystallogr E Crystallogr Commun 2023; 79:895-898. [PMID: 37817954 PMCID: PMC10561198 DOI: 10.1107/s2056989023007624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/01/2023] [Indexed: 10/12/2023]
Abstract
The asymmetric unit of the title compound, C12H13N3O4, consists of two mol-ecules differing to a small degree in their conformations. In the crystal, layers of mol-ecules are connected by weak C-H⋯O hydrogen bonds and slipped π-stacking inter-actions. These layers lie parallel to (10) and are stacked along the normal to that plane. Hirshfeld surface analysis indicates that the most important contributions for the crystal packing arise from H⋯H (43.5%) and H⋯O/O⋯H (30.8%) contacts. The density functional theory (DFT) optimized structure of the title compound at the B3LYP/ 6-311 G(d,p) level agrees well with the experimentally determined mol-ecular structure in the solid state.
Collapse
Affiliation(s)
- Seqqat Yousra
- Laboratory of Applied Organic Chemistry, Faculty of Science and Technology, Sidi Mohammed Ben Abdullah University, Route d’Immouzzer, BP 2202, Fez, Morocco
| | - Lhoussaine El Ghayati
- Laboratory of Heterocyclic Organic Chemistry, Medicines Science Research Center, Pharmacochemistry Competence Center, Mohammed V University in Rabat, Faculty of Sciences, Morocco
| | - Tuncer Hökelek
- Department of Physics, Hacettepe University, 06800 Beytepe, Ankara, Türkiye
| | - Fouad Ouazzani Chahdi
- Laboratory of Applied Organic Chemistry, Faculty of Science and Technology, Sidi Mohammed Ben Abdullah University, Route d’Immouzzer, BP 2202, Fez, Morocco
| | - Joel T. Mague
- Department of Chemistry, Tulane University, New Orleans, LA 70118, USA
| | - Youssef Kandri Rodi
- Laboratory of Applied Organic Chemistry, Faculty of Science and Technology, Sidi Mohammed Ben Abdullah University, Route d’Immouzzer, BP 2202, Fez, Morocco
| | - Nada Kheira Sebbar
- Laboratory of Heterocyclic Organic Chemistry, Medicines Science Research Center, Pharmacochemistry Competence Center, Mohammed V University in Rabat, Faculty of Sciences, Morocco
- Laboratory of Organic and physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, Ibn Zohr University, Agadir, Morocco
| |
Collapse
|
6
|
Okoro CO, Fatoki TH. A Mini Review of Novel Topoisomerase II Inhibitors as Future Anticancer Agents. Int J Mol Sci 2023; 24:ijms24032532. [PMID: 36768852 PMCID: PMC9916523 DOI: 10.3390/ijms24032532] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Several reviews of inhibitors of topoisomerase II have been published, covering research before 2018. Therefore, this review is focused primarily on more recent publications with relevant points from the earlier literature. Topoisomerase II is an established target for anticancer drugs, which are further subdivided into poisons and catalytic inhibitors. While most of the topoisomerase II-based drugs in clinical use are mostly topoisomerase II poisons, their mechanism of action has posed severe concern due to DNA damaging potential, including the development of multi-drug resistance. As a result, we are beginning to see a gradual paradigm shift towards non-DNA damaging agents, such as the lesser studied topoisomerase II catalytic inhibitors. In addition, this review describes some novel selective catalytic topoisomerase II inhibitors. The ultimate goal is to bring researchers up to speed by curating and delineating new scaffolds as the leads for the optimization and development of new potent, safe, and selective agents for the treatment of cancer.
Collapse
|
7
|
Avula B, Reddivari CKR, Muchumarri RMR, Eraganaboyina S, Zyryanov GV, Nemallapudi BR. Recent Advances in the Synthesis of Quinoxalines. A Mini Review. Polycycl Aromat Compd 2023. [DOI: 10.1080/10406638.2023.2167215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Balakrishna Avula
- Department of Chemistry, Rajeev Gandhi Memorial College of Engineering and Technology (Autonomous), Nandyal, India
| | | | | | - Suneetha Eraganaboyina
- Department of Chemistry, Rajeev Gandhi Memorial College of Engineering and Technology (Autonomous), Nandyal, India
| | - Grigory V. Zyryanov
- Chemical Engineering Institute, Ural Federal University, Yekaterinburg, Russia
- Ural Division of the Russian Academy of Sciences, I. Ya. Postovskiy Institute of Organic Synthesis, Yekaterinburg, Russia
| | | |
Collapse
|
8
|
Protein-ligand binding affinity prediction with edge awareness and supervised attention. iScience 2022; 26:105892. [PMID: 36691617 PMCID: PMC9860494 DOI: 10.1016/j.isci.2022.105892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/12/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Accurate prediction of protein-ligand binding affinity is crucial in structure-based drug design but remains some challenges even with recent advances in deep learning: (1) Existing methods neglect the edge information in protein and ligand structure data; (2) current attention mechanisms struggle to capture true binding interactions in the small dataset. Herein, we proposed SEGSA_DTA, a SuperEdge Graph convolution-based and Supervised Attention-based Drug-Target Affinity prediction method, where the super edge graph convolution can comprehensively utilize node and edge information and the multi-supervised attention module can efficiently learn the attention distribution consistent with real protein-ligand interactions. Results on the multiple datasets show that SEGSA_DTA outperforms current state-of-the-art methods. We also applied SEGSA_DTA in repurposing FDA-approved drugs to identify potential coronavirus disease 2019 (COVID-19) treatments. Besides, by using SHapley Additive exPlanations (SHAP), we found that SEGSA_DTA is interpretable and further provides a new quantitative analytical solution for structure-based lead optimization.
Collapse
|
9
|
Elwan A, Sakr H, El-Helby AGA, El-morsy A, Abdelgawad MA, Ghoneim MM, El-Sherbiny M, El-Adl K. Triazoloquinoxalines-based DNA intercalators-Topo II inhibitors: design, synthesis, docking, ADMET and anti-proliferative evaluations. J Enzyme Inhib Med Chem 2022; 37:1556-1567. [PMID: 35635148 PMCID: PMC9154796 DOI: 10.1080/14756366.2022.2080205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Affiliation(s)
- Alaa Elwan
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Helmy Sakr
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Abdel-Ghany A. El-Helby
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed El-morsy
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, Faculty of Pharmacy, AlMaarefa University, Ad Diriyah, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Khaled El-Adl
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| |
Collapse
|
10
|
Asahara H, Bonkohara A, Takagi M, Iwai K, Ito A, Yoshioka K, Tani S, Umezu K, Nishiwaki N. Development of a synthetic equivalent of α,α-dicationic acetic acid leading to unnatural amino acid derivatives via tetrafunctionalized methanes. Org Biomol Chem 2022; 20:2282-2292. [PMID: 35234775 DOI: 10.1039/d1ob02482e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diethyl mesoxalate (DEMO) exhibits high electrophilicity and accepts the nucleophilic addition of a less nucleophilic acid amide to afford N,O-hemiacetal. However, our research showed that elimination of the amide moiety proceeded more easily than dehydration upon treatment with a base. This problem was overcome by reacting DEMO with an acid amide in the presence of acetic anhydride to efficiently obtain N,O-acetal. Acetic acid was eliminated leading to the formation of N-acylimine in situ upon treatment with the base. N-Acylimine is also electrophilic, accepting the second nucleophilic addition by pyrrole or indole to form α,α-disubstituted malonates. Subsequent hydrolysis followed by decarboxylation resulted in (α-indolyl-α-acylamino)acetic acid formation; homologs of tryptophan. Through this process, DEMO serves as a synthetic equivalent of α,α-dicationic acetic acid to facilitate nucleophilic introduction of the two substituents.
Collapse
Affiliation(s)
- Haruyasu Asahara
- School of Environmental Science and Engineering, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan. .,Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka 1-6, Suita, Osaka 565-0871, Japan
| | - Atsushi Bonkohara
- School of Environmental Science and Engineering, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan.
| | - Masaya Takagi
- School of Environmental Science and Engineering, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan.
| | - Kento Iwai
- School of Environmental Science and Engineering, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan. .,Research Center for Molecular Design, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan
| | - Akitaka Ito
- School of Environmental Science and Engineering, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan. .,Research Center for Molecular Design, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan
| | - Kotaro Yoshioka
- Kumiai Chemical Industry Co. Ltd., Nakanogo, Fuji, Shizuoka 421-3306, Japan
| | - Shinki Tani
- Kumiai Chemical Industry Co. Ltd., Nakanogo, Fuji, Shizuoka 421-3306, Japan
| | - Kazuto Umezu
- Kumiai Chemical Industry Co. Ltd., Nakanogo, Fuji, Shizuoka 421-3306, Japan
| | - Nagatoshi Nishiwaki
- School of Environmental Science and Engineering, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan. .,Research Center for Molecular Design, Kochi University of Technology, Tosayamada, Kami, Kochi 782-8502, Japan
| |
Collapse
|
11
|
El-Adl K, Ibrahim MK, Alesawy MS, Eissa IH. Triazoloquinazoline derived classical DNA intercalators: Design, synthesis, in silico ADME profile, docking, and antiproliferative evaluations. Arch Pharm (Weinheim) 2022; 355:e2100506. [PMID: 35293628 DOI: 10.1002/ardp.202100506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 11/08/2022]
Abstract
Thirteen novel [1,2,4]triazolo[4,3-c]quinazoline derivatives as DNA intercalators were synthesized and their anticancer activities evaluated against HepG2 and HCT-116 cells. A docking study was carried out to explore how the new derivatives bind to active sites of DNA. The docking data were highly interrelated with that of biological testing. The HCT-116 cell line was the most sensitive one to the effect of the new derivatives. Compound 7c exhibited the highest anticancer activities against both the HepG2 and HCT116 cancer cell lines. Despite this compound displaying less activity than doxorubicin, it could be useful as a template for future manipulation, optimization, and investigation to produce other analogs with potential activity. The most active derivatives, 7c , 7b , and 7a were evaluated as DNA binders. Compound 7c displayed the highest binding affinity. Furthermore, the absorption, distribution, metabolism, excretion, and toxicity (ADMET) profile was calculated for the four most active compounds in comparison to doxorubicin as reference drug. Our derivatives 7a , 7b , and 7c displayed a very good calculated ADMET profile in comparison to doxorubicin.
Collapse
Affiliation(s)
- Khaled El-Adl
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Al-Azhar University, Cairo, Egypt.,Pharmaceutical Chemistry Department, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Mohamed-Kamal Ibrahim
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Al-Azhar University, Cairo, Egypt
| | - Mohamed S Alesawy
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Al-Azhar University, Cairo, Egypt
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
12
|
Eissa IH, Ibrahim MK, Alesawy MS, El-Adl K. Antiproliferative evaluations of triazoloquinazolines as classical DNA intercalators: Design, synthesis, ADMET profile, and molecular docking. Arch Pharm (Weinheim) 2022; 355:e2100487. [PMID: 35194810 DOI: 10.1002/ardp.202100487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 01/08/2023]
Abstract
Novel triazoloquinazolines were designed and synthesized and evaluated as anticancer agents against HepG2 and HCT-116 cells. The biological testing data corresponded well to those of the molecular docking studies. The HCT-116 cell line was most affected due to the actions of our derivatives. Derivative 7a was the most potent one against both HepG2 and HCT116 cells, with IC50 = 7.98 and 5.57 µM, respectively. This compound showed anticancer activity that was nearly equipotent to that of doxorubicin against HepG2 cells, but higher than that of doxorubicin against HCT116 cells (IC50 = 7.94 and 8.07 µM, respectively). Compounds 8, 7b , and 6f showed excellent anticancer activities against both the HCT116 and HepG2 cell lines. The highly active compounds 6f , 7a , 7b , and 8 were evaluated for their DNA-binding activities. Compounds 7a and 8 showed the highest binding activities. These derivatives potently intercalate in DNA, at IC50 values of 42.90 and 48.13 µM, respectively. Derivatives 6f and 7b showed good DNA-binding activities, with IC50 values of 54.24 and 50.56 µM, respectively. Furthermore, in silico calculated ADMET profiles were established for our four highly active derivatives, in comparison to doxorubicin. Our derivatives 6f , 7a , 7b , and 8 showed a very good ADMET profile. Compounds 6f , 7a , 7b , and 8 follow Lipinski's rules, while doxorubicin violates three of these rules.
Collapse
Affiliation(s)
- Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohamed-Kamal Ibrahim
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohamed S Alesawy
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Khaled El-Adl
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt.,Chemistry Department, Faculty of Pharmacy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| |
Collapse
|
13
|
Alesawy MS, Ibrahim M, Eissa IH, El‐Adl K. Design, synthesis, in silico ADMET, docking, and antiproliferative evaluations of [1,2,4]triazolo[4,3‐
c
]quinazolines as classical DNA intercalators. Arch Pharm (Weinheim) 2022; 355:e2100412. [DOI: 10.1002/ardp.202100412] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Mohamed S. Alesawy
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy Al‐Azhar University Cairo Egypt
| | - Mohamed‐Kamal Ibrahim
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy Al‐Azhar University Cairo Egypt
| | - Ibrahim H. Eissa
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy Al‐Azhar University Cairo Egypt
| | - Khaled El‐Adl
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy Al‐Azhar University Cairo Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy Heliopolis University for Sustainable Development Cairo Egypt
| |
Collapse
|
14
|
Zhao Y, Aziz AUR, Zhang H, Zhang Z, Li N, Liu B. A systematic review on active sites and functions of PIM-1 protein. Hum Cell 2022; 35:427-440. [PMID: 35000143 DOI: 10.1007/s13577-021-00656-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
The Proviral Integration of Molony murine leukemia virus (PIM)-1 protein contributes to the solid cancers and hematologic malignancies, cell growth, proliferation, differentiation, migration, and other life activities. Many studies have related these functions to its molecular structure, subcellular localization and expression level. However, recognition of specific active sites and their effects on the activity of this constitutively active kinase is still a challenge. Based on the close relationship between its molecular structure and functional activity, this review covers the specific residues involved in the binding of ATP and different substrates in its catalytic domain. This review then elaborates on the relevant changes in protein conformation and cell functions after PIM-1 binds to different substrates. Therefore, this intensive study can improve the understanding of PIM-1-regulated signaling pathways by facilitating the discovery of its potential phosphorylation substrates.
Collapse
Affiliation(s)
- Youyi Zhao
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Aziz Ur Rehman Aziz
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Panjin Campus of Dalian University of Technology, Panjin, 124221, China
| | - Na Li
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China.
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
15
|
Dibenzofuran Derivatives Inspired from Cercosporamide as Dual Inhibitors of Pim and CLK1 Kinases. Molecules 2021; 26:molecules26216572. [PMID: 34770981 PMCID: PMC8587151 DOI: 10.3390/molecules26216572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022] Open
Abstract
Pim kinases (proviral integration site for Moloney murine leukemia virus kinases) are overexpressed in various types of hematological malignancies and solid carcinomas, and promote cell proliferation and survival. Thus, Pim kinases are validated as targets for antitumor therapy. In this context, our combined efforts in natural product-inspired library generation and screening furnished very promising dibenzo[b,d]furan derivatives derived from cercosporamide. Among them, lead compound 44 was highlighted as a potent Pim-1/2 kinases inhibitor with an additional nanomolar IC50 value against CLK1 (cdc2-like kinases 1) and displayed a low micromolar anticancer potency towards the MV4-11 (AML) cell line, expressing high endogenous levels of Pim-1/2 kinases. The design, synthesis, structure-activity relationship, and docking studies are reported herein and supported by enzyme, cellular assays, and Galleria mellonella larvae testing for acute toxicity.
Collapse
|
16
|
Alsaif NA, Taghour MS, Alanazi MM, Obaidullah AJ, Alanazi WA, Alasmari A, Albassam H, Dahab MA, Mahdy HA. Identification of new [1,2,4]triazolo[4,3-a]quinoxalines as potent VEGFR-2 tyrosine kinase inhibitors: Design, synthesis, anticancer evaluation, and in silico studies. Bioorg Med Chem 2021; 46:116384. [PMID: 34479065 DOI: 10.1016/j.bmc.2021.116384] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
Tumor angiogenesis is mainly regulated by VEGFR-2. In this study, a new series of [1,2,4]triazolo[4,3-a]quinoxaline based-derivatives has been designed and synthesized to develop new anti-proliferative and anti-VEGFR-2 members. Anti-proliferative activities of the synthesized compounds were tested against MCF-7 and HepG2 cell lines. Compound 19a exhibited the highest activity towards both MCF-7 and HepG2 cell lines (IC50 = 8.2 and 5.4 µM, respectively), compared to sorafenib (IC50 = 3.51 and 2.17 µM, respectively). Additionally, all compounds were screened to evaluate their effect as VEGFR-2 inhibitors. Compound 19a (IC50 = 3.4 nM) exhibited good activity compared to sorafenib (IC50 = 3.12 nM). Furthermore, compound 19a disrupted the HepG2 cell cycle by arresting the G2/M phase. Also, marked increase in the percentage apoptotic cells was achieved by compound 19a. The induced apoptotic effect of compound 19a in HepG2 cells was assured by increased pro-apoptotic marker (Bax) expression by 2.33-fold and decreased anti-apoptotic (Bcl-2) expression by 1.88-fold, resulting in an elevation of the Bax/Bcl-2 ratio in HepG2 cells. Comparing to the control cells, compound 19a induced an increase in expression of cleaved caspase-3 and caspase-9 by 2.44- and 2.69-fold, respectively. Finally, the binding modes of the target derivatives were investigated through docking studies against the proposed molecular target (VEGFR-2, PDB ID: 2OH4).
Collapse
Affiliation(s)
- Nawaf A Alsaif
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed S Taghour
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wael A Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11541, Saudi Arabia
| | - Abdullah Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11541, Saudi Arabia
| | - Hussam Albassam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11541, Saudi Arabia
| | - Mohammed A Dahab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Hazem A Mahdy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| |
Collapse
|
17
|
Yi X, Cao Z, Yuan Y, Li W, Cui X, Chen Z, Hu X, Yu A. Design and synthesis of a novel mitochondria-targeted osteosarcoma theranostic agent based on a PIM1 kinase inhibitor. J Control Release 2021; 332:434-447. [PMID: 33662457 DOI: 10.1016/j.jconrel.2021.02.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022]
Abstract
Osteosarcoma (OS) is the most common malignancy of the skeletal system, with a poor prognosis and high rate of recurrence. Adequate surgical margin and adjuvant chemotherapy improve the overall survival and limb salvage rate of osteosarcoma patients. Previous studies have showed that OS exhibits an increase in the expression of proviral integration site for Moloney murine leukemia virus 1 (PIM1) kinase, and high levels of PIM1 are also associated with poor OS prognosis and metastasis. We exploited the overexpression of proto-oncogenic PIM1 in OS towards the development of a novel near-infrared imaging and targeted therapeutic agent, namely QCAi-Cy7d by conjugating a PIM1 small molecule inhibitor and heptamethine cyanine dye, for simultaneous guiding surgery and chemotherapy. QCAi-Cy7d showed targeted imaging and anticancer activities against OS in vitro and vivo without any obvious toxicity, and its antitumoral activity was much greater than the parent PIMI inhibitor. These results demonstrated the potential of new conjugate of PIM1 inhibitor and near-infrared imaging, supporting structure-based design and development of theranostic agents for precise tumor imaging and targeted treatment.
Collapse
Affiliation(s)
- Xinzeyu Yi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhi Cao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Ying Yuan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wen Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Xinyue Cui
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Zilin Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China.
| | - Xiang Hu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
18
|
New Quinoxaline Derivatives as Dual Pim-1/2 Kinase Inhibitors: Design, Synthesis and Biological Evaluation. Molecules 2021; 26:molecules26040867. [PMID: 33562106 PMCID: PMC7914722 DOI: 10.3390/molecules26040867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 11/17/2022] Open
Abstract
Proviral integration site for Moloney murine leukemia virus (Pim)-1/2 kinase overexpression has been identified in a variety of hematologic (e.g., multiple myeloma or acute myeloid leukemia (AML)) and solid (e.g., colorectal carcinoma) tumors, playing a key role in cancer progression, metastasis, and drug resistance, and is linked to poor prognosis. These kinases are thus considered interesting targets in oncology. We report herein the design, synthesis, structure–activity relationships (SAR) and in vitro evaluations of new quinoxaline derivatives, acting as dual Pim1/2 inhibitors. Two lead compounds (5c and 5e) were then identified, as potent submicromolar Pim-1 and Pim-2 inhibitors. These molecules were also able to inhibit the growth of the two human cell lines, MV4-11 (AML) and HCT-116 (colorectal carcinoma), expressing high endogenous levels of Pim-1/2 kinases.
Collapse
|
19
|
El-Adl K, Ibrahim MK, Alesawy MS, Eissa IH. [1,2,4]Triazolo[4,3-c]quinazoline and bis([1,2,4]triazolo)[4,3-a:4′,3′-c]quinazoline derived DNA intercalators: Design, synthesis, in silico ADMET profile, molecular docking and anti-proliferative evaluation studies. Bioorg Med Chem 2021; 30:115958. [DOI: 10.1016/j.bmc.2020.115958] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023]
|
20
|
El-Adl K, El-Helby AGA, Sakr H, Elwan A. [1,2,4]Triazolo[4,3-a]quinoxaline and [1,2,4]triazolo[4,3-a]quinoxaline-1-thiol-derived DNA intercalators: design, synthesis, molecular docking, in silico ADMET profiles and anti-proliferative evaluations. NEW J CHEM 2021. [DOI: 10.1039/d0nj02990d] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In view of their DNA intercalation activities as anticancer agents, 17 novel [1,2,4]triazolo[4,3-a]quinoxaline derivatives have been designed, synthesized and evaluated against HepG2, HCT-116 and MCF-7 cells.
Collapse
Affiliation(s)
- Khaled El-Adl
- Pharmaceutical Chemistry Department
- Faculty of Pharmacy
- Al-Azhar University
- Cairo
- Egypt
| | | | - Helmy Sakr
- Pharmaceutical Chemistry Department
- Faculty of Pharmacy
- Al-Azhar University
- Cairo
- Egypt
| | - Alaa Elwan
- Pharmaceutical Chemistry Department
- Faculty of Pharmacy
- Al-Azhar University
- Cairo
- Egypt
| |
Collapse
|
21
|
El-Adl K, El-Helby AGA, Sakr H, Elwan A. Design, synthesis, molecular docking and anti-proliferative evaluations of [1,2,4]triazolo[4,3-a]quinoxaline derivatives as DNA intercalators and Topoisomerase II inhibitors. Bioorg Chem 2020; 105:104399. [DOI: 10.1016/j.bioorg.2020.104399] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/27/2020] [Accepted: 10/17/2020] [Indexed: 11/15/2022]
|
22
|
Shyamsivappan S, Saravanan A, Vivek R, Suresh T, Shankar R, Gothandam KM, Mohan PS. Novel phenyl and thiophene dispiro indenoquinoxaline pyrrolidine quinolones induced apoptosis via G1/S and G2/M phase cell cycle arrest in MCF-7 cells. NEW J CHEM 2020. [DOI: 10.1039/d0nj02588g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
New phenyl and thiophene dispiro indeno quinoxaline pyrrolidine quinolone analogues were synthesized by a one-pot four-component [3+2] cycloaddition reaction between (E)-3-arylidene-2,3-dihydro-8-nitro-4-quinolones, o-phenylenediamine, ninhydrin, and benzylamine/thiophenemethylamine.
Collapse
Affiliation(s)
| | | | - Raju Vivek
- Cancer Research Program (CRP)
- Bio-Nano Therapeutics Research Laboratory
- School of Life Sciences
- Department of Zoology
- Bharathiar University
| | | | | | - K. M. Gothandam
- School of Bio-Sciences and Technology
- Vellore Institute of Technology
- Vellore
- India
| | | |
Collapse
|
23
|
Eissa IH, Metwaly AM, Belal A, Mehany ABM, Ayyad RR, El-Adl K, Mahdy HA, Taghour MS, El-Gamal KMA, El-Sawah ME, Elmetwally SA, Elhendawy MA, Radwan MM, ElSohly MA. Discovery and antiproliferative evaluation of new quinoxalines as potential DNA intercalators and topoisomerase II inhibitors. Arch Pharm (Weinheim) 2019; 352:e1900123. [PMID: 31463953 DOI: 10.1002/ardp.201900123] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022]
Abstract
In continuation of our previous work on the design and synthesis of topoisomerase II (Topo II) inhibitors and DNA intercalators, a new series of quinoxaline derivatives were designed and synthesized. The synthesized compounds were evaluated for their cytotoxic activities against a panel of three cancer cell lines (Hep G-2, Hep-2, and Caco-2). Compounds 18b, 19b, 23, 25b, and 26 showed strong potencies against all tested cell lines with IC50 values ranging from 0.26 ± 0.1 to 2.91 ± 0.1 µM, comparable with those of doxorubicin (IC50 values ranging from 0.65 ± 0.1 to 0.81 ± 0.1 µM). The most active compounds were further evaluated for their Topo II inhibitory activities and DNA intercalating affinities. Compounds 19b and 19c exhibited high activities against Topo II (IC50 = 0.97 ± 0.1 and 1.10 ± 0.1 µM, respectively) and bound the DNA at concentrations of 43.51 ± 2.0 and 49.11 ± 1.8 µM, respectively, whereas compound 28b exhibited a significant affinity to bind the DNA with an IC50 value of 37.06 ± 1.8 µM. Moreover, apoptosis and cell-cycle tests of the most promising compound 19b were carried out. It was found that 19b can significantly induce apoptosis in Hep G-2 cells. It has revealed cell-cycle arrest at the G2/M phase. Moreover, compound 19b downregulated the Bcl-2 levels, indicating its potential to enhance apoptosis. Furthermore, molecular docking studies were carried out against the DNA-Topo II complex to examine the binding patterns of the synthesized compounds.
Collapse
Affiliation(s)
- Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed M Metwaly
- Department of Pharmacognosy, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Amany Belal
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.,Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmed B M Mehany
- Department of Zoology, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Rezk R Ayyad
- Pharmaceutical Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Khaled El-Adl
- Pharmaceutical Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Drug Technology, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Hazem A Mahdy
- Pharmaceutical Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohammed S Taghour
- Pharmaceutical Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Kamal M A El-Gamal
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohamad E El-Sawah
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Souad A Elmetwally
- Department of Basic Science, Higher Technological Institute, 10th of Ramadan City, Egypt
| | - Mostafa A Elhendawy
- National Center for Natural Products Research, University of Mississippi, Mississippi
| | - Mohamed M Radwan
- National Center for Natural Products Research, University of Mississippi, Mississippi.,Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, University of Mississippi, Mississippi.,Department of Pharmaceutics and Drug Delivery, University of Mississippi, Mississippi, Mississippi
| |
Collapse
|
24
|
Jaiswal D, Tiwari J, Singh S, Sharma AK, Singh J, Singh J. Rose Bengal Catalyzed Coupling of 1, 2 ‐ Dicarbonyls and Phenylene 1, 2 ‐Diamines: Visible‐Light Mediated Synthesis of Quinoxalines. ChemistrySelect 2019. [DOI: 10.1002/slct.201902080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Deepali Jaiswal
- Environmentally Benign Synthesis LabDepartment of ChemistryUniversity of Allahabad Allahabad- 211002 India
| | - Jyoti Tiwari
- Environmentally Benign Synthesis LabDepartment of ChemistryUniversity of Allahabad Allahabad- 211002 India
| | - Shailesh Singh
- Environmentally Benign Synthesis LabDepartment of ChemistryUniversity of Allahabad Allahabad- 211002 India
| | - Amit Kumar Sharma
- Environmentally Benign Synthesis LabDepartment of ChemistryUniversity of Allahabad Allahabad- 211002 India
| | - Jaya Singh
- Department of ChemistryLRPG College, Sahibabad Ghaziabad- 201005 India
| | - Jagdamba Singh
- Environmentally Benign Synthesis LabDepartment of ChemistryUniversity of Allahabad Allahabad- 211002 India
| |
Collapse
|
25
|
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]
|
26
|
Virtual screening identification of novel chemical inhibitors for aberrant interactions between pathogenic mutant SOD1 and tubulin. Neurochem Int 2019; 126:19-26. [PMID: 30831216 DOI: 10.1016/j.neuint.2019.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/22/2019] [Accepted: 02/25/2019] [Indexed: 01/26/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal neurodegenerative disease caused by selective motor neuron death. Mutations in the gene encoding copper/zinc superoxide dismutase (SOD1) belong to one of the four major mutation clusters responsible for pathogenesis of ALS. Toxic gain-of-function (not loss-of-function) of SOD1 mutants causes motor neuron degeneration. Aberrant protein-protein interactions (PPI) between mutant SOD1 and other proteins are involved in this toxic gain-of-function. Therefore, PPI inhibitors of mutant SOD1 not only increase understanding of ALS pathogenesis, but can also be used as novel therapeutics for ALS. Although it is challenging to identify PPI inhibitors, prior knowledge of the binding site can increase success probability. We have previously reported that tubulin interacts with N-terminal residues 1-23 of mutant SOD1. In the present study, we performed virtual screening by docking simulation of 32,791 compounds using this N-terminal binding site as prior knowledge. An established assay system for interaction inhibition between mutant SOD1-tubulin was used as an in-house model system to identify mutant SOD1 PPI inhibitors, with the goal of developing novel therapeutics for ALS. Consequently, five of six assay-executable compounds among top-ranked compounds during docking simulation inhibited the mutant SOD1-tubulin interaction in vitro. Binding mode analysis predicted that some inhibitors might bind the tubulin binding site of G85R SOD1 by pi electron interaction with the aromatic ring of the Trp32 residue of G85R SOD1. Our screening methods may contribute to the identification of lead compounds for treating ALS.
Collapse
|
27
|
Ding RR, Yuan JL, Jia YN, Liao XM, Wang SS, Shao ZM, Feng MY, Jie W, Shen ZH. Epstein-Barr virus-encoded LMP1 regulated Pim1 kinase expression promotes nasopharyngeal carcinoma cells proliferation. Onco Targets Ther 2019; 12:1137-1146. [PMID: 30809095 PMCID: PMC6376889 DOI: 10.2147/ott.s190274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Epstein–Barr virus-encoded LMP1 plays a critical role in the carcinogenesis of nasopharyngeal carcinoma (NPC), but the mechanism remains elusive. We aimed to analyze the expression and clinical pathological significance of provirus integration site for Moloney murine leukemia virus 1 (Pim1) in clinical NPC, and to elucidate the effect of LMP1 on Pim1 expression and its mechanism. Methods Immunohistochemical staining was used to detect the expression of Pim1 in clinical NPC tissues and control nasopharyngeal chronic inflammation (NPI) tissues, and the correlation between Pim1 and clinical parameters of NPC patients was analyzed. The LMP1 stable expression cell line CNE1-LMP1-OV was constructed through infecting the well-differentiated nasopharyngeal carcinoma cells CNE1 with LMP1 overexpressing lentivirus. Then the in vivo experiments were conducted. Results Among 89 NPC patients, 48 cases (53.93%) were positive for Pim1, while only one case was Pim1 positive in 15 NPI controls (6.67%). Pim1 expression was not correlated with gender, age, smoking status and clinical classification of NPC patients, but positively correlated with T, N and M classification. CNE1-LMP1-OV cell line was successfully established, which displayed a higher cell proliferation ability and Pim1 expression. NF-κB inhibitor PDTC, PKC inhibitor GF109203X and STAT3 inhibitor Stattic significantly attenuated LMP1-induced Pim1 expression, and while AP-1 inhibitor SR11302 showed no inhibitory effect. Interestingly, Pim1 inhibitor quercetagetin significantly inhibited the proliferation of CNE1-LMP1-OV cells. Conclusion LMP1 mediates Pim1 expression through NF-κB, PKC and STAT3 signaling, which promotes the proliferation of NPC cells and participate in the clinical progression of NPC.
Collapse
Affiliation(s)
- Ran-Ran Ding
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Jian-Ling Yuan
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Ya-Nan Jia
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China, .,Department of Pathology, Dongguan People's Hospital, Dongguan 523059, China
| | - Xiao-Min Liao
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Si-Si Wang
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Zhong-Ming Shao
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Mu-Yin Feng
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Wei Jie
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| | - Zhi-Hua Shen
- Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, China,
| |
Collapse
|