1
|
Merwid-Ląd A, Ziółkowski P, Nowak B, Świątek P, Szczukowski Ł, Kwiatkowska J, Piasecka K, Szeląg A, Szandruk-Bender M. 1,3,4-Oxadiazole Derivatives of Pyrrolo[3,4- d]pyridazinone Alleviate TNBS-Induced Colitis and Exhibit No Significant Testicular Toxicity. Pharmaceuticals (Basel) 2025; 18:546. [PMID: 40283981 PMCID: PMC12030013 DOI: 10.3390/ph18040546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/26/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Inflammatory bowel disease significantly impairs the patient's quality of life. In young individuals, both the disease and the drugs used for the treatment may impact fertility. Our study aimed to assess the action of new 1,3,4-oxadiazole derivatives of pyrrolo[3,4-d]pyridazinone on the rat testes in a model of TNBS-induced colitis in rats. Methods: In the current study, testes from eight randomly chosen rats were taken from each of the following groups: the control group (K), the colitis group (C), and the groups receiving compounds 7b, 10b, and 13b in higher doses (20 mg/kg). Results: Colitis did not affect the testicular index (expressed as a percentage of the body weight), but in group 13b, this parameter was significantly higher than in group K. No significant differences between groups were noticed in malondialdehyde, superoxide dismutase, interleukin-1, or metalloproteinase 9 levels. In the colitis group, lactate dehydrogenase activity in the testes was not increased; however, the administration of compound 10b significantly increased this parameter when compared to both groups K and C. Histological evaluation also did not reveal abnormalities, and the morphology of the testicular tissues was comparable in all groups. Conclusions: The results may suggest that the new 1,3,4-oxadiazole derivatives of pyrrolo[3,4-d]pyridazinone did not exert significant testicular toxicity.
Collapse
Affiliation(s)
- Anna Merwid-Ląd
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.N.); (A.S.); (M.S.-B.)
| | - Piotr Ziółkowski
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, Marcinkowskiego 1, 50-368 Wrocław, Poland;
| | - Beata Nowak
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.N.); (A.S.); (M.S.-B.)
| | - Piotr Świątek
- Department of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland; (P.Ś.); (Ł.S.)
| | - Łukasz Szczukowski
- Department of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland; (P.Ś.); (Ł.S.)
| | - Joanna Kwiatkowska
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.N.); (A.S.); (M.S.-B.)
| | - Katarzyna Piasecka
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.N.); (A.S.); (M.S.-B.)
| | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.N.); (A.S.); (M.S.-B.)
| | - Marta Szandruk-Bender
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.N.); (A.S.); (M.S.-B.)
| |
Collapse
|
2
|
Saadh MJ, Ahmed HH, Kareem RA, Jain V, Ballal S, Singh A, Sharma GC, Devi A, Nasirov A, Sameer HN, Yaseen A, Athab ZH, Adil M. In Silico design and molecular dynamics analysis of imidazole derivatives as selective cyclooxygenase-2 inhibitors. Comput Biol Chem 2025; 115:108341. [PMID: 39808951 DOI: 10.1016/j.compbiolchem.2025.108341] [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: 12/08/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Cyclooxygenase-2 (COX-2), a key enzyme in the inflammatory pathway, is the target for various nonsteroidal anti-inflammatory drugs (NSAIDs) and selective inhibitors known as coxibs. This study focuses on the development of novel imidazole derivatives as COX-2 inhibitors, utilizing a Structure-Activity Relationship (SAR) approach to enhance binding affinity and selectivity. Molecular docking was performed using Autodock Vina, revealing binding energies of -6.928, -7.187, and -7.244 kJ/mol for compounds 5b, 5d, and 5e, respectively. Molecular dynamics simulations using GROMACS provided insights into the stability and conformational changes of the protein-ligand complexes. Key metrics such as RMSD, RMSF, Rg, SASA, and hydrogen bond analysis were employed to assess the interactions. The binding free energy of the inhibitors was estimated using the MMPBSA method, highlighting compound 5b (N-[(3-benzyl-2-methylsulfonylimidazol-4-yl)methyl]-4-methoxyaniline) with the lowest binding energy of -162.014 kcal/mol. ADMET analysis revealed that compound 5b exhibited the most favorable pharmacokinetic properties and safety profile. Overall, this investigation underscores the potential of these novel imidazole derivatives as effective COX-2 inhibitors, with compound 5b emerging as the most promising candidate for further development.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Vicky Jain
- Marwadi University Research Center, Department of Chemistry, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Abdulaziz Nasirov
- Department of Psychiatry, narcology and pediatric narcology, medical psychology and psychotherapy, Tashkent Pediatric Medical Institute, Bogishamol Street 223, Tashkent 100140, Uzbekistan
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
3
|
El-Mahdy RY, Galal N, Lotfy R, Arafa RK. Structure-based design of new anticancer N3-Substituted quinazolin-4-ones as type I ATP-competitive inhibitors targeting the deep hydrophobic pocket of EGFR. Comput Biol Med 2025; 186:109640. [PMID: 39765103 DOI: 10.1016/j.compbiomed.2024.109640] [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: 10/04/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 02/20/2025]
Abstract
Epidermal growth factor receptor (EGFR) is amongst the earliest targeted kinases by small-molecule inhibitors for the management of EGFR-positive cancer types. While a few inhibitors are granted FDA approval for clinical use, discovery of new inhibitors is still of merit to enhance ligand-binding stability and subsequent enzyme inhibition. Thus, a structure-based design approach was adopted to devise a new series of twenty-nine N3-substituted quinazolin-4-ones as type I ATP-competitive inhibitors targeting the deep hydrophobic pocket of EGFR. The most active compounds demonstrated potent IC50s against MDA-MB-231 and HepG2 cancer cells being comparable to or better than the reference drugs erlotinib and lapatinib. IC50s of 5f and 15a against MDA-MB-231 were 3.34 and 4.99, whilst those against HepG2 were 6.37 and 2.18 μM, respectively. Also, members of this series demonstrated selective cytotoxicity against cancer cell lines showing low toxicity on human skin fibroblast normal cells hFB-4. Both 5f and 15a also effectively inhibited EGFR with sub-micromolar respective IC50s of 0.07 and 0.12 μM. The two derivatives halted the cell cycle progression of treated cancer cells and induced apoptosis as affirmed by flow cytometry along with RT-PCR-determined overexpression of the pro-apoptotic genes p53, Caspase 3, and Bax. Notably, docking and molecular dynamics simulations of members of this series of quinazolin-4-one derivatives showed that analogs with a short linker at the N3 position of the quinazoline ring exemplified by 5f bind to the active form of EGFR with their terminal aryl ring dwelling in the BPI pocket similar to erlotinib, while those with a longer linker represented by 15a bind to the inactive form in a comparable manner to lapatinib lodging the terminal phenyl in the BPII pocket.
Collapse
Affiliation(s)
- Ragaa Y El-Mahdy
- Drug Design and Discovery Lab, Helmy Institute of Medical Sciences, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt
| | - Noha Galal
- Drug Design and Discovery Lab, Helmy Institute of Medical Sciences, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt
| | - Rahma Lotfy
- Drug Design and Discovery Lab, Helmy Institute of Medical Sciences, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt
| | - Reem K Arafa
- Drug Design and Discovery Lab, Helmy Institute of Medical Sciences, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science, Technology and Innovation, Giza, 12578, Egypt.
| |
Collapse
|
4
|
Gaber AA, El-Morsy AM, Sherbiny FF, Bayoumi AH, El-Gamal KM, El-Adl K, Al-Karmalawy AA, Ezz Eldin RR, Saleh MA, Abulkhair HS. Pharmacophore-linked pyrazolo[3,4-d]pyrimidines as EGFR-TK inhibitors: Synthesis, anticancer evaluation, pharmacokinetics, and in silico mechanistic studies. Arch Pharm (Weinheim) 2025; 358:e2100258. [PMID: 34467546 DOI: 10.1002/ardp.202100258] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023]
Abstract
Targeting the epidermal growth factor receptors (EGFRs) with small inhibitor molecules has been validated as a potential therapeutic strategy in cancer therapy. Pyrazolo[3,4-d]pyrimidine is a versatile scaffold that has been exploited for developing potential anticancer agents. On the basis of fragment-based drug discovery, considering the essential pharmacophoric features of potent EGFR tyrosine kinase (TK) inhibitors, herein, we report the design and synthesis of new hybrid molecules of the pyrazolo[3,4-d]pyrimidine scaffold linked with diverse pharmacophoric fragments with reported anticancer potential. These fragments include hydrazone, indoline-2-one, phthalimide, thiourea, oxadiazole, pyrazole, and dihydropyrazole. The synthesized molecules were evaluated for their anticancer activity against the human breast cancer cell line, MCF-7. The obtained results revealed comparable antitumor activity with that of the reference drugs doxorubicin and toceranib. Docking studies were performed along with EGFR-TK and ADMET profiling studies. The results of the docking studies showed the ability of the designed compounds to interact with key residues of the EGFR-TK through a number of covalent and noncovalent interactions. The obtained activity of compound 25 (IC50 = 2.89 µM) suggested that it may serve as a lead for further optimization and drug development.
Collapse
Affiliation(s)
- Ahmed A Gaber
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
| | - Ahmed M El-Morsy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Farag F Sherbiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Department of Chemistry, Basic Science Center and Pharmaceutical Organic Chemistry College of Pharmaceutical Science & Drug Manufacturing, Misr University for Science and Technology (MUST), Al-Motamayez District, 6th of October City, Egypt
| | - Ashraf H Bayoumi
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
| | - Kamal M El-Gamal
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
| | - Khaled El-Adl
- Department of Medicinal Chemistry & Drug Design, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| | - Rogy R Ezz Eldin
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Marwa A Saleh
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| |
Collapse
|
5
|
Mahboubi-Rabbani M, Abdolghaffari AH, Ghesmati M, Amini A, Zarghi A. Selective COX-2 inhibitors as anticancer agents: a patent review (2018-2023). Expert Opin Ther Pat 2024; 34:733-757. [PMID: 38958471 DOI: 10.1080/13543776.2024.2373771] [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: 12/11/2023] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION COX-2 is a crucial enzyme in the manufacture of prostaglandins. The enzyme's metabolites might have an important function as regulators of the inflammatory response and other medical conditions such as cancer. Selective COX-2 inhibitors are believed to enhance or reverse the response of cancer chemotherapeutics. AREAS COVERED This study addresses the chemical structures as well as the antitumor activity of new COX-2 inhibitors produced in the recent five years, aiming to provide an insight into the mechanism of COX-2 induced PGE2 powerful signal in cancer development. EXPERT OPINION The significance of selective COX-2 inhibitors as an efficient superfamily of compounds with anti-inflammatory, anti-Alzheimer's, anti-Parkinson's disease, and anticancer properties has piqued the passion of academics in the field of drug development. Long-term usage of selective COX-2 inhibitors, such as celecoxib has been proven in clinical trials to lower the incidence of several human malignancies. Furthermore, celecoxib has the potential to greatly increase the effectiveness of chemotherapy. Our extensive understanding of selective COX-2 inhibitor SAR may aid in the development of safer and more effective selective COX-2 inhibitors as cancer chemopreventive agents. This review focuses on the different structural classes of selective COX-2 inhibitors, with a particular emphasis on their SAR.
Collapse
Affiliation(s)
- Mohammad Mahboubi-Rabbani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Ghesmati
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Amini
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Zarghi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zhang J, Wang Z, Wang J, Zhuo X, Yu L, Han T, Song Y, Gai C, Zou Y, Meng Q, Chai X, Zhao Q. Total synthesis and structural modification of the dibenzylbutane lignan LCA as a potent anti-inflammatory agent against LPS-induced acute lung injury. Eur J Med Chem 2024; 268:116272. [PMID: 38402749 DOI: 10.1016/j.ejmech.2024.116272] [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: 08/29/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Acute lung injury (ALI) is a serious public health problem associated with high morbidity and mortality. However, few efficacious drugs are clinically available. Inhibition of proinflammatory cytokines is considered to be a promising method for the treatment of inflammatory diseases. Herein, the total synthesis of a dibenzylbutane lignan, 9'-O-di-(E)-feruloyl-meso-5,5'-dimethoxysecoisolariciresinol (LCA), was completed. A series of LCA derivatives were designed and synthesized, and their anti-inflammatory activities were evaluated. Derivative 14r significantly inhibited LPS-induced expression of NO and the proinflammatory cytokines TNF-α, IL-6, and IL-1β in RAW 264.7 cells and inhibited activation of the NF-κB pathway. Compound 14r reduced LPS-induced pulmonary inflammation and ALI in mice. It showed significant protective effects against LPS-induced ALI in mice and significantly reduced levels of proinflammatory cytokines in serum and bronchoalveolar lavage fluid. The ratio of wet weight to dry weight of lung tissue was normalized by compound 14r, which was consistent with suppression of neutrophil infiltration and production of proinflammatory cytokines. Compound 14r reduced the mRNA expression of some proinflammatory cytokines, improved histopathologic changes, and reduced macrophage infiltration in lung tissues. Collectively, these results suggest a new series of LCA derivatives that could be promising anti-inflammatory agents for ALI treatment.
Collapse
Affiliation(s)
- Juan Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Zhen Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Jing Wang
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Xiaobin Zhuo
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Luyao Yu
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Ting Han
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yan Song
- Navy Medical Center, Second Military Medical University, Shanghai, 200433, China
| | - Conghao Gai
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yan Zou
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| | - Xiaoyun Chai
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Qingjie Zhao
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
7
|
Wang A, Shuai W, Wu C, Pei J, Yang P, Wang X, Li S, Liu J, Wang Y, Wang G, Ouyang L. Design, Synthesis, and Biological Evaluation of Dual Inhibitors of EGFR L858R/T790M/ACK1 to Overcome Osimertinib Resistance in Nonsmall Cell Lung Cancers. J Med Chem 2024; 67:2777-2801. [PMID: 38323982 DOI: 10.1021/acs.jmedchem.3c01934] [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: 02/08/2024]
Abstract
Activation of the alternative pathways and abnormal signaling transduction are frequently observed in third-generation EGFR-TKIs (epidermal growth factor receptor tyrosine kinase inhibitors)-resistant patients. Wherein, hyperphosphorylation of ACK1 contributes to EGFR-TKIs acquired resistance. Dual inhibition of EGFRL858R/T790M and ACK1 might improve therapeutic efficacy and overcome resistance in lung cancers treatment. Here, we identified a EGFRL858R/T790M/ACK1 dual-targeting compound 21a with aminoquinazoline scaffold, which showed excellent inhibitory activities against EGFRL858R/T790M (IC50 = 23 nM) and ACK1 (IC50 = 263 nM). The cocrystal and docking analysis showed that 21a occupied the ATP binding pockets of EGFRL858R/T790M and ACK1. Moreover, 21a showed potent antiproliferative activities against the H1975 cells, MCF-7 cells and osimertinib-resistant cells AZDR. Further, 21a showed significant antitumor effects and good safety in ADZR xenograft-bearing mice. Taken together, 21a was a potent dual inhibitor of EGFRL858R/T790M/ACK1, which is deserved as a potential lead for overcoming acquired resistance to osimertinib during the EGFR-targeted therapy.
Collapse
Affiliation(s)
- Aoxue Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Chengyong Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Panpan Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xin Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Shutong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Jiaxi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yuxi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Alam MM, Alsenani NI, Abdelhamid AA, Ahmad A, Baothman OA, Hosawi SA, Altayeb H, Nadeem MS, Ahmad V, Nazreen S, Elhenawy AA. New paracetamol hybrids as anticancer and COX-2 inhibitors: Synthesis, biological evaluation and docking studies. Arch Pharm (Weinheim) 2024; 357:e2300340. [PMID: 37880869 DOI: 10.1002/ardp.202300340] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/31/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023]
Abstract
Drug repurposing is an emerging field in drug development that has provided many successful drugs. In the current study, paracetamol, a known antipyretic and analgesic agent, was chemically modified to generate paracetamol derivatives as anticancer and anticyclooxygenase-2 (COX-2) agents. Compound 11 bearing a fluoro group was the best cytotoxic candidate with half-maximal inhibitory concentration (IC50 ) values ranging from 1.51 to 6.31 μM and anti-COX-2 activity with IC50 = 0.29 μM, compared to the standard drugs, doxorubicin and celecoxib. The cell cycle and apoptosis studies revealed that compound 11 possesses the ability to induce cell cycle arrest in the S phase and apoptosis in colon Huh-7 cells. These results were strongly supported by docking studies, which showed strong interactions with the amino acids of the COX-2 protein, and in silico pharmacokinetic predictions were found to be favorable for these newly synthesized paracetamol derivatives. It can be concluded that compound 11 could block cell growth and proliferation by inhibiting the COX-2 enzyme in cancer therapy.
Collapse
Affiliation(s)
- Mohammad Mahboob Alam
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Nawaf I Alsenani
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Antar A Abdelhamid
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Abrar Ahmad
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Othman A Baothman
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Salman A Hosawi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hisham Altayeb
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammad Shahid Nadeem
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Varish Ahmad
- Department of Health Information Technology, Faculty of Applied Studies, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Syed Nazreen
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Ahmed A Elhenawy
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
9
|
Zhang J, Wang Z, Gai C, Yang F, Yun X, Jiang B, Zou Y, Meng Q, Zhao Q, Chai X. Design, synthesis, evaluation and optimization of novel azole analogues as potent antifungal agents. Bioorg Med Chem 2024; 97:117543. [PMID: 38071944 DOI: 10.1016/j.bmc.2023.117543] [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: 09/23/2023] [Revised: 11/13/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
In order to develop antifungal drugs, a series of novel azole analogues were designed and synthesized based on our previous work. Most of the target compounds had broad-spectrum antifungal activity, which showed excellent to moderate inhibitory activity against the tested strains, except A. fum 0504656. Among these, compounds B3, B7, B8, B11, B12 and E9 showed excellent activity against C. alb Y0109 and C. alb SC5314 (with the MIC80: 0.0156 ug/mL). In addition, compound B3 showed the best inhibitory activity against fluconazole-resistant strains C. alb 901 and C. alb 904, and had low toxicity against NIH/3T3 cells at the effective MIC range against fungi. Structure-activity relationship and docking studies of the derivatives suggest that the presence of the 2-fluoro-4-hydroxyphenyl and 1,2,3-triazole group enhance the antifungal activity of the compounds, which may be related to the interaction of the key groups with the amino acids surrounding the target enzyme.
Collapse
Affiliation(s)
- Juan Zhang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Zhen Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Conghao Gai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Fan Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Xiaoqing Yun
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Boye Jiang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yan Zou
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Qingjie Zhao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Xiaoyun Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
10
|
Chaudhary T, Upadhyay PK. A Bird's Eye Review of Recent Reports on 1,3,4-oxadiazoles' Anti-inflammatory Insights Perspectives. Curr Org Synth 2024; 21:595-606. [PMID: 37157211 DOI: 10.2174/1570179420666230508124847] [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: 11/24/2022] [Revised: 03/08/2023] [Accepted: 03/22/2023] [Indexed: 05/10/2023]
Abstract
Anti-inflammatory agents suppress inflammatory mediators such as prostaglandins, prostacyclins, cytokines, thromboxane, histamine, bradykinins, COX-I and COX-II, 5-LOX, and other substances. These inflammatory chemicals create inflammatory responses when tissue is injured by trauma, bacteria, heat, toxins, or other factors. These inflammatory reactions may result in fluid flow from the blood vessels into the tissues, resulting in swelling. When the therapeutic importance of these clinically beneficial medications in treating inflammation was recognized, it spurred the invention of even more powerful and important molecules. Oxadiazole derivatives are exceptionally potent NSAIDs, and they are widely used. Comprehensive biochemical, structure-activity-relationship and pharmacological investigations have demonstrated that these 1,3,4-oxadiazole compounds exhibit anti-inflammatory properties. This review article outlines the synthesis scheme for 1,3,4-oxadiazole used in treating inflammation.
Collapse
Affiliation(s)
- Tarun Chaudhary
- Institute of Pharmaceutical Research, Department of Chemistry, GLA University, Mathura, Uttar Pradesh, India
| | - Prabhat Kumar Upadhyay
- Institute of Pharmaceutical Research, Department of Chemistry, GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
11
|
Wang C, Zhang Y, Zhang T, Xu J, Yan S, Liang B, Xing D. Epidermal growth factor receptor dual-target inhibitors as a novel therapy for cancer: A review. Int J Biol Macromol 2023; 253:127440. [PMID: 37839594 DOI: 10.1016/j.ijbiomac.2023.127440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
Overexpression of the epidermal growth factor receptor (EGFR) has been linked to several human cancers, including esophageal cancer, pancreatic cancer, anal cancer, breast cancer, and lung cancer, particularly non-small cell lung cancer (NSCLC). Therefore, EGFR has emerged as a critical target for treating solid tumors. Many 1st-, 2nd-, 3rd-, and 4th-generation EGFR single-target inhibitors with clinical efficacy have been designed and synthesized in recent years. Drug resistance caused by EGFR mutations has posed a significant challenge to the large-scale clinical application of EGFR single-target inhibitors and the discovery of novel EGFR inhibitors. Therapeutic methods for overcoming multipoint EGFR mutations are still needed in medicine. EGFR dual-target inhibitors are more promising than single-target inhibitors as they have a lower risk of drug resistance, higher efficacy, lower dosage, and fewer adverse events. EGFR dual-target inhibitors have been developed sequentially to date, providing new options for remission in patients with previously untreatable malignancies and laying the groundwork for a future generation of compounds. This paper introduces the EGFR family proteins and their synergistic effects with other anticancer targets, and provides a comprehensive review of the development of EGFR dual-target inhibitors in cancer, as well as the opportunities and challenges associated with those fields.
Collapse
Affiliation(s)
- Chao Wang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao 266071, Shandong, China.
| | - Tingting Zhang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China
| | - Saisai Yan
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China.
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
12
|
Zhang F, Zhu G, Li Y, Qi Y, Wang Z, Li W. Dual-target inhibitors based on COX-2: a review from medicinal chemistry perspectives. Future Med Chem 2023; 15:2209-2233. [PMID: 38095081 DOI: 10.4155/fmc-2023-0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/08/2023] [Indexed: 12/20/2023] Open
Abstract
Inhibitors of COX-2 constitute a class of anti-inflammatory analgesics, showing potential against certain types of cancer. However, such inhibitors are associated with cardiovascular toxicity. Moreover, although single-target molecules possess specificity for particular targets, they often lead to poor safety, low efficacy and drug resistance due to compensatory mechanisms. A new generation of dual-target drugs that simultaneously inhibit COX-2 and another target is showing strong potential to treat cancer or reduce adverse cardiac effects. The present perspective focuses on the structure and functions of COX-2, and its role as a therapeutic target. It also explores the current state and future possibilities for dual-target strategies from a medicinal chemistry perspective.
Collapse
Affiliation(s)
- Fengmei Zhang
- Department of Pulmonary & Critical Care Medicine, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Respiratory Health & Multimorbidity, West China Hospital, Chengdu, 610041, Sichuan, China
| | - Guonian Zhu
- Department of Pulmonary & Critical Care Medicine, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Respiratory Health & Multimorbidity, West China Hospital, Chengdu, 610041, Sichuan, China
| | - Yangqian Li
- Department of Pulmonary & Critical Care Medicine, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Respiratory Health & Multimorbidity, West China Hospital, Chengdu, 610041, Sichuan, China
| | - Yawen Qi
- Department of Pulmonary & Critical Care Medicine, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Respiratory Health & Multimorbidity, West China Hospital, Chengdu, 610041, Sichuan, China
| | - Zhoufeng Wang
- Department of Pulmonary & Critical Care Medicine, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Respiratory Health & Multimorbidity, West China Hospital, Chengdu, 610041, Sichuan, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, 610041, Sichuan, China
| | - Weimin Li
- Department of Pulmonary & Critical Care Medicine, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Respiratory Health & Multimorbidity, West China Hospital, Chengdu, 610041, Sichuan, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, 610041, Sichuan, China
| |
Collapse
|
13
|
Ali A, Wani AB, Malla BA, Poyya J, Dar NJ, Ali F, Ahmad SB, Rehman MU, Nadeem A. Network Pharmacology Integrated Molecular Docking and Dynamics to Elucidate Saffron Compounds Targeting Human COX-2 Protein. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2058. [PMID: 38138161 PMCID: PMC10744988 DOI: 10.3390/medicina59122058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: Cyclooxygenase-2 (COX-2) is mostly linked to inflammation and has been validated as a molecular target for treating inflammatory diseases. The present study aimed to identify novel compounds that could inhibit COX-2, which is associated with various diseases including inflammation, and in such a scenario, plant-derived biomolecules have been considered as attractive candidates. Materials and Methods: In the present study, physiochemical properties and toxicity of natural compounds/drugs were determined by SWISSADME and ProTox-II. In the present study, the molecular docking binding features of saffron derivatives (crocetin, picrocrocin, quercetin, safranal, crocin, rutin, and dimethylcrocetin) against human COX-2 protein were assessed. Moreover, protein-protein interactions, topographic properties, gene enrichment analysis and molecular dynamics simulation were also determined. Results: The present study revealed that picrocrocin showed the highest binding affinity of -8.1 kcal/mol when docked against the COX-2 protein. PROCHECK analysis revealed that 90.3% of the protein residues were found in the most favored region. Compartmentalized Protein-Protein Interaction identified 90 interactions with an average interaction score of 0.62, and the highest localization score of 0.99 found in secretory pathways. The Computed Atlas of Surface Topography of Proteins was used to identify binding pockets and important residues that could serve as drug targets. Use of WEBnmα revealed protein dynamics by using normal mode analysis. Ligand and Receptor Dynamics used the Molecular Generalized Born Surface Area approach to determine the binding free energy of the protein. Gene enrichment analysis revealed that ovarian steroidogenesis, was the most significant enrichment pathway. Molecular dynamic simulations were executed for the best docked (COX-2-picrocrocin) complex, and the results displayed conformational alterations with more pronounced surface residue fluctuations in COX-2 with loss of the intra-protein hydrogen bonding network. The direct interaction of picrocrocin with various crucial amino-acid residues like GLN203, TYR385, HIS386 and 388, ASN382, and TRP387 causes modifications in these residues, which ultimately attenuates the activity of COX-2 protein. Conclusions: The present study revealed that picrocrocin was the most effective biomolecule and could be repurposed via computational approaches. However, various in vivo and in vitro observations are still needed.
Collapse
Affiliation(s)
- Aarif Ali
- Division of Veterinary Biochemistry, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K, Shuhama, Alusteng, Srinagar 190006, India
| | - Amir Bashir Wani
- Genome Engineering and Societal Biotechnology Lab., Division of Plant Biotechnology, SKUAST-K, Shalimar, Srinagar 190006, India;
| | - Bashir Ahmad Malla
- Department of Biochemistry, School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar 190006, India;
| | - Jagadeesha Poyya
- SDM Research Institute for Biomedical Sciences, Dharwad 580009, India
| | - Nawab John Dar
- SALK Institute for Biological Studies, La Jolla, San Diego, CA 92037, USA;
| | - Fasil Ali
- Department of Studies and Research in Biochemistry, Mangalore University, Mangalore 571232, India
| | - Sheikh Bilal Ahmad
- Division of Veterinary Biochemistry, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-K, Shuhama, Alusteng, Srinagar 190006, India
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
14
|
Chahal S, Rani P, Kiran, Sindhu J, Joshi G, Ganesan A, Kalyaanamoorthy S, Mayank, Kumar P, Singh R, Negi A. Design and Development of COX-II Inhibitors: Current Scenario and Future Perspective. ACS OMEGA 2023; 8:17446-17498. [PMID: 37251190 PMCID: PMC10210234 DOI: 10.1021/acsomega.3c00692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/21/2023] [Indexed: 09/29/2023]
Abstract
Innate inflammation beyond a threshold is a significant problem involved in cardiovascular diseases, cancer, and many other chronic conditions. Cyclooxygenase (COX) enzymes are key inflammatory markers as they catalyze prostaglandins production and are crucial for inflammation processes. While COX-I is constitutively expressed and is generally involved in "housekeeping" roles, the expression of the COX-II isoform is induced by the stimulation of different inflammatory cytokines and also promotes the further generation of pro-inflammatory cytokines and chemokines, which affect the prognosis of various diseases. Hence, COX-II is considered an important therapeutic target for drug development against inflammation-related illnesses. Several selective COX-II inhibitors with safe gastric safety profiles features that do not cause gastrointestinal complications associated with classic anti-inflammatory drugs have been developed. Nevertheless, there is mounting evidence of cardiovascular side effects from COX-II inhibitors that resulted in the withdrawal of market-approved anti-COX-II drugs. This necessitates the development of COX-II inhibitors that not only exhibit inhibit potency but also are free of side effects. Probing the scaffold diversity of known inhibitors is vital to achieving this goal. A systematic review and discussion on the scaffold diversity of COX inhibitors are still limited. To address this gap, herein we present an overview of chemical structures and inhibitory activity of different scaffolds of known COX-II inhibitors. The insights from this article could be helpful in seeding the development of next-generation COX-II inhibitors.
Collapse
Affiliation(s)
- Sandhya Chahal
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Payal Rani
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Kiran
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Jayant Sindhu
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Gaurav Joshi
- Department
of Pharmaceutical Sciences, Hemvati Nandan
Bahuguna Garhwal (A Central) University, Chauras Campus, Tehri Garhwal, Uttarakhand 249161, India
- Adjunct
Faculty at Department of Biotechnology, Graphic Era (Deemed to be) University, 566/6, Bell Road, Clement Town, Dehradun, Uttarakhand 248002, India
| | - Aravindhan Ganesan
- ArGan’sLab,
School of Pharmacy, University of Waterloo, Waterloo, Ontario N2G 1C5, Canada
| | | | - Mayank
- University
College of Pharmacy, Guru Kashi University, Talwandi Sabo, Punjab 151302, India
| | - Parvin Kumar
- Department
of Chemistry, Kurukshetra University, Kurukshetra 136119, India
| | - Rajvir Singh
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 02150, Finland
| |
Collapse
|
15
|
Kumar D, Aggarwal N, Deep A, Kumar H, Chopra H, Marwaha RK, Cavalu S. An Understanding of Mechanism-Based Approaches for 1,3,4-Oxadiazole Scaffolds as Cytotoxic Agents and Enzyme Inhibitors. Pharmaceuticals (Basel) 2023; 16:254. [PMID: 37259401 PMCID: PMC9963071 DOI: 10.3390/ph16020254] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 07/30/2023] Open
Abstract
The world's health system is plagued by cancer and a worldwide effort is underway to find new drugs to treat cancer. There has been a significant improvement in understanding the pathogenesis of cancer, but it remains one of the leading causes of death. The imperative 1,3,4-oxadiazole scaffold possesses a wide variety of biological activities, particularly for cancer treatment. In the development of novel 1,3,4-oxadiazole-based drugs, structural modifications are important to ensure high cytotoxicity towards malignant cells. These structural modification strategies have shown promising results when combined with outstanding oxadiazole scaffolds, which selectively interact with nucleic acids, enzymes, and globular proteins. A variety of mechanisms, such as the inhibition of growth factors, enzymes, and kinases, contribute to their antiproliferative effects. The activity of different 1,3,4-oxadiazole conjugates were tested on the different cell lines of different types of cancer. It is demonstrated that 1,3,4-oxadiazole hybridization with other anticancer pharmacophores have different mechanisms of action by targeting various enzymes (thymidylate synthase, HDAC, topoisomerase II, telomerase, thymidine phosphorylase) and many of the proteins that contribute to cancer cell proliferation. The focus of this review is to highlight the anticancer potential, molecular docking, and SAR studies of 1,3,4-oxadiazole derivatives by inhibiting specific cancer biological targets, such as inhibiting telomerase activity, HDAC, thymidylate synthase, and the thymidine phosphorylase enzyme. The purpose of this review is to summarize recent developments and discoveries in the field of anticancer drugs using 1,3,4-oxadiazoles.
Collapse
Affiliation(s)
- Davinder Kumar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Navidha Aggarwal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Aakash Deep
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani 127021, India
| | - Harsh Kumar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India
| | - Rakesh Kumar Marwaha
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
16
|
Mohsin NUA, Aslam S, Ahmad M, Irfan M, Al-Hussain SA, Zaki MEA. Cyclooxygenase-2 (COX-2) as a Target of Anticancer Agents: A Review of Novel Synthesized Scaffolds Having Anticancer and COX-2 Inhibitory Potentialities. Pharmaceuticals (Basel) 2022; 15:ph15121471. [PMID: 36558921 PMCID: PMC9783503 DOI: 10.3390/ph15121471] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a serious threat to human beings and is the second-largest cause of death all over the globe. Chemotherapy is one of the most common treatments for cancer; however, drug resistance and severe adverse effects are major problems associated with anticancer therapy. New compounds with multi-target inhibitory properties are targeted to surmount these challenges. Cyclooxygenase-2 (COX-2) is overexpressed in cancers of the pancreas, breast, colorectal, stomach, and lung carcinoma. Therefore, COX-2 is considered a significant target for the synthesis of new anticancer agents. This review discusses the biological activity of recently prepared dual anticancer and COX-2 inhibitory agents. The most important intermolecular interactions with the COX-2 enzyme have also been presented. Analysis of these agents in the active area of the COX-2 enzyme could guide the introduction of new lead compounds with extreme selectivity and minor side effects.
Collapse
Affiliation(s)
- Noor ul Amin Mohsin
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sana Aslam
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.A.); (M.E.A.Z.)
| | - Muhammad Irfan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
- Correspondence: (M.A.); (M.E.A.Z.)
| |
Collapse
|
17
|
Cell Cycle Arrest and Apoptosis-Inducing Ability of Benzimidazole Derivatives: Design, Synthesis, Docking, and Biological Evaluation. Molecules 2022; 27:molecules27206899. [PMID: 36296495 PMCID: PMC9607330 DOI: 10.3390/molecules27206899] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 12/02/2022] Open
Abstract
In the current study, new benzimidazole-based 1,3,4-oxadiazole derivatives have been synthesized and characterized by NMR, IR, MS, and elemental analysis. The final compounds were screened for cytotoxicity against MDA-MB-231, SKOV3, and A549 cell lines and EGFR for inhibitory activities. Compounds 10 and 13 were found to be the most active against all the tested cell lines, comparable to doxorubicin, and exhibited significant inhibition on EGFR kinase, with IC50 0.33 and 0.38 μM, respectively, comparable to erlotinib (IC50 0.39 μM). Furthermore, these two compounds effectively suppressed cell cycle progression and induced cell apoptosis in MDA-MB-231, SKOV3, and A549 cell lines. The docking studies revealed that these compounds showed interactions similar to erlotinib at the EGFR site. It can be concluded that the synthesized molecules effectively inhibit EGFR, can arrest the cell cycle, and may trigger apoptosis and therefore, could be used as lead molecules in the development of new anticancer agents targeting EGFR kinase.
Collapse
|
18
|
Abdelhaleem EF, Kassab AE, El-Nassan HB, Khalil OM. Design, synthesis, and biological evaluation of new celecoxib analogs as apoptosis inducers and cyclooxygenase-2 inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200190. [PMID: 35976138 DOI: 10.1002/ardp.202200190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022]
Abstract
Series of new celecoxib analogs were synthesized to assess their anticancer activity against the MCF-7 cell line. Four compounds, 3a, 3c, 5b, and 5c, showed 1.4-9.2-fold more potent anticancer activity than celecoxib. The antiproliferative activity of the most potent compounds, 3c, 5b, and 5c, seems to be associated well with their ability to induce apoptosis in MCF-7 cells (18-24-fold). This evidence was supported by an increase in the expression of the tumor suppressor gene p53 (4-6-fold), the elevation in the Bax/BCL-2 ratio, and a significant increase in the level of active caspase-7 (4-7-fold). Moreover, compounds 3c and 5c showed significant cyclooxygenase-2 (COX-2) inhibitory activity. They were also docked into the crystal structure of the COX-2 enzyme (PDB ID: 3LN1) to understand their mode of binding.
Collapse
Affiliation(s)
- Eman F Abdelhaleem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala B El-Nassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Omneya M Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
19
|
Design, synthesis and mechanistic studies of novel imidazo[1,2-a]pyridines as anticancer agents. Bioorg Chem 2022; 128:106042. [PMID: 35878430 DOI: 10.1016/j.bioorg.2022.106042] [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: 03/20/2022] [Revised: 05/16/2022] [Accepted: 07/17/2022] [Indexed: 11/20/2022]
Abstract
Herein, the design, synthesis and mechanistic study of five series of imidazo[1,2-a]pyridines 8a-d, 9a-f, 11a-c, 12a-d and 14a-d as anticancer agents were discussed. The cytotoxicity of imidazo[1,2-a]pyridine derivatives was screened against NCI 60 cancer cell lines. The cytotoxicity of compounds 8b, 8c, 9e and 9f was then evaluated against leukemia K-562 cancer cell line and normal lung fibroblasts (WI38). The hydrazone derivatives 8b and 8c exhibited significant cytotoxic activities against the leukemia K-562 cancer cell line with good safety margins (IC50 = 2.91 µM, SI = 8.32 and IC50 = 1.09 µM, SI = 10.54, respectively). In addition, compounds 8b, 8c, 9e and 9f were tested for their EGFR and COX-2 inhibitory activities. The hydrazone derivatives 8b and 8c were the most active EGFR inhibitors with IC50 values of 0.123 and 0.072 µM, respectively. Compound 8c selectively inhibited COX-2 (IC50 = 1.09 µM, SI = 13.78). Moreover, the potential of compound 8c to induce apoptosis in leukemia K-562 cell line was determined. Compound 8c showed a pre-G1 apoptosis and a growth arrest of leukemia K-562 cell line at G1 phase of cell cycle. Also, compound 8c was able to induce caspase-3 overexpression (6.98 folds), if compared to control. Finally, molecular docking studies and physicochemical properties calculation of compounds 8b, 8c, 9e and 9f were carried out to explain the biological data and to predict bioavailability of the most active compounds.
Collapse
|
20
|
Shi K, Wang G, Pei J, Zhang J, Wang J, Ouyang L, Wang Y, Li W. Emerging strategies to overcome resistance to third-generation EGFR inhibitors. J Hematol Oncol 2022; 15:94. [PMID: 35840984 PMCID: PMC9287895 DOI: 10.1186/s13045-022-01311-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/28/2022] [Indexed: 02/08/2023] Open
Abstract
Epidermal growth factor receptor (EGFR), the receptor for members of the epidermal growth factor family, regulates cell proliferation and signal transduction; moreover, EGFR is related to the inhibition of tumor cell proliferation, angiogenesis, invasion, metastasis, and apoptosis. Therefore, EGFR has become an important target for the treatment of cancer, including non-small cell lung cancer, head and neck cancer, breast cancer, glioma, cervical cancer, and bladder cancer. First- to third-generation EGFR inhibitors have shown considerable efficacy and have significantly improved disease prognosis. However, most patients develop drug resistance after treatment. The challenge of overcoming intrinsic and acquired resistance in primary and recurrent cancer mediated by EGFR mutations is thus driving the search for alternative strategies in the design of new therapeutic agents. In view of resistance to third-generation inhibitors, understanding the intricate mechanisms of resistance will offer insight for the development of more advanced targeted therapies. In this review, we discuss the molecular mechanisms of resistance to third-generation EGFR inhibitors and review recent strategies for overcoming resistance, new challenges, and future development directions.
Collapse
Affiliation(s)
- Kunyu Shi
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.,Tianfu Jincheng Laboratory, Chengdu, 610041, China
| | - Guan Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Junping Pei
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.,Tianfu Jincheng Laboratory, Chengdu, 610041, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Liang Ouyang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China. .,Tianfu Jincheng Laboratory, Chengdu, 610041, China.
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Tianfu Jincheng Laboratory, Chengdu, 610041, China.
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Tianfu Jincheng Laboratory, Chengdu, 610041, China.
| |
Collapse
|
21
|
Bakchi B, Krishna AD, Sreecharan E, Ganesh VBJ, Niharika M, Maharshi S, Puttagunta SB, Sigalapalli DK, Bhandare RR, Shaik AB. An overview on applications of SwissADME web tool in the design and development of anticancer, antitubercular and antimicrobial agents: A medicinal chemist's perspective. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
22
|
Exploring the composition of Syringa reticulata subsp. amurensis seed and its underlying mechanism against chronic bronchitis. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2022. [DOI: 10.1016/j.cjac.2022.100132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Ruslin R, Yamin Y, Kasmawati H, Mangrura S, Kadidae L, Alroem A, Arba M. The Search for Cyclooxygenase-2 (COX-2) Inhibitors for the Treatment of Inflammation Disease: An in-silico Study. J Multidiscip Healthc 2022; 15:783-791. [PMID: 35444425 PMCID: PMC9014226 DOI: 10.2147/jmdh.s359429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/22/2022] [Indexed: 01/26/2023] Open
Affiliation(s)
- Ruslin Ruslin
- Faculty of Pharmacy, Universitas Halu Oleo, Kendari, 93232, Indonesia
- Correspondence: Ruslin Ruslin, Email
| | - Yamin Yamin
- Faculty of Pharmacy, Universitas Halu Oleo, Kendari, 93232, Indonesia
| | - Henny Kasmawati
- Faculty of Pharmacy, Universitas Halu Oleo, Kendari, 93232, Indonesia
| | - Samuel Mangrura
- Department of Chemistry, Faculty of Mathematics and Natural; Sciences, Universitas Halu Oleo, Kendari, 93232, Indonesia
| | - Laode Kadidae
- Department of Chemistry, Faculty of Mathematics and Natural; Sciences, Universitas Halu Oleo, Kendari, 93232, Indonesia
| | - Armid Alroem
- Department of Chemistry, Faculty of Mathematics and Natural; Sciences, Universitas Halu Oleo, Kendari, 93232, Indonesia
| | - Muhammad Arba
- Faculty of Pharmacy, Universitas Halu Oleo, Kendari, 93232, Indonesia
| |
Collapse
|
24
|
Hu L, Fan M, Shi S, Song X, Wang F, He H, Qi B. Dual target inhibitors based on EGFR: Promising anticancer agents for the treatment of cancers (2017-). Eur J Med Chem 2022; 227:113963. [PMID: 34749202 DOI: 10.1016/j.ejmech.2021.113963] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023]
Abstract
The EGFR family play a significant role in cell signal transduction and their overexpression is implicated in the pathogenesis of numerous human solid cancers. Inhibition of the EGFR-mediated signaling pathways by EGFR inhibitors is a widely used strategy for the treatment of cancers. In most cases, the EGFR inhibitors used in clinic were only effective when the cancer cells harbored specific activating EGFR mutations which appeared to preserve the ligand-dependency of receptor activation but altered the pattern of downstream signaling pathways. Moreover, cancer is a kind of multifactorial disease, and therefore manipulating a single target may result in treatment failure. Although drug combinations for the treatment of cancers proved to be successful, the use of two or more drugs concurrently still was a challenge in clinical therapy owing to various dose-limiting toxicities and drug-drug interactions caused by pharmacokinetic profiles changed. Therefore, a single drug targeting two or multiple targets could serve as an effective strategy for the treatment of cancers. In recent, drugs with diverse pharmacological effects have been shown to be more advantageous than combination therapies due to their lower incidences of side effects and more resilient therapies. Accordingly, dual target-single-agent strategy has become a popular field for cancer treatment, and researchers became more and more interest in the development of novel dual-target drugs in recent years. In this review, we briefly introduce the EGFR family proteins and synergisms between EGFR and other anticancer targets, and summarizes the development of potential dual target inhibitors based on wild-type and/or mutant EGFR for the treatment of solid cancers in the past five years. Additionally, the rational design and SARs of these dual target agents are also presented in detailed, which will lay a significant foundation for the further development of novel EGFR-based dual inhibitors with excellent druggability.
Collapse
Affiliation(s)
- Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| | - Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| |
Collapse
|
25
|
Recent development on COX-2 inhibitors as promising anti-inflammatory agents: The past 10 years. Acta Pharm Sin B 2022; 12:2790-2807. [PMID: 35755295 PMCID: PMC9214066 DOI: 10.1016/j.apsb.2022.01.002] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/14/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Cyclooxygenases play a vital role in inflammation and are responsible for the production of prostaglandins. Two cyclooxygenases are described, the constitutive cyclooxygenase-1 and the inducible cyclooxygenase-2, for which the target inhibitors are the non-steroidal anti-inflammatory drugs (NSAIDs). Prostaglandins are a class of lipid compounds that mediate acute and chronic inflammation. NSAIDs are the most frequent choices for treatment of inflammation. Nevertheless, currently used anti-inflammatory drugs have become associated with a variety of adverse effects which lead to diminished output even market withdrawal. Recently, more studies have been carried out on searching novel selective COX-2 inhibitors with safety profiles. In this review, we highlight the various structural classes of organic and natural scaffolds with efficient COX-2 inhibitory activity reported during 2011–2021. It will be valuable for pharmaceutical scientists to read up on the current chemicals to pave the way for subsequent research.
Collapse
|
26
|
New N-Substituted-1,2,4-triazole Derivatives of Pyrrolo[3,4- d]pyridazinone with Significant Anti-Inflammatory Activity-Design, Synthesis and Complementary In Vitro, Computational and Spectroscopic Studies. Int J Mol Sci 2021; 22:ijms222011235. [PMID: 34681894 PMCID: PMC8540742 DOI: 10.3390/ijms222011235] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 01/05/2023] Open
Abstract
Regarding that the chronic use of commonly available non-steroidal and anti-inflammatory drugs (NSAIDs) is often restricted by their adverse effects, there is still a current need to search for and develop new, safe and effective anti-inflammatory agents. As a continuation of our previous work, we designed and synthesized a series of 18 novel N-substituted-1,2,4-triazole-based derivatives of pyrrolo[3,4-d]pyridazinone 4a-c-9a-c. The target compounds were afforded via a convenient way of synthesis, with good yields. The executed cell viability assay revealed that molecules 4a-7a, 9a, 4b-7b, 4c-7c do not exert a cytotoxic effect and were qualified for further investigations. According to the performed in vitro test, compounds 4a-7a, 9a, 4b, 7b, 4c show significant cyclooxygenase-2 (COX-2) inhibitory activity and a promising COX-2/COX-1 selectivity ratio. These findings are supported by a molecular docking study which demonstrates that new derivatives take position in the active site of COX-2 very similar to Meloxicam. Moreover, in the carried out in vitro evaluation within cells, the title molecules increase the viability of cells pre-incubated with the pro-inflammatory lipopolysaccharide and reduce the level of reactive oxygen and nitrogen species (RONS) in induced oxidative stress. The spectroscopic and molecular modeling study discloses that new compounds bind favorably to site II(m) of bovine serum albumin. Finally, we have also performed some in silico pharmacokinetic and drug-likeness predictions. Taking all of the results into consideration, the molecules belonging to series a (4a-7a, 9a) show the most promising biological profile.
Collapse
|
27
|
Baykov SV, Mikherdov AS, Novikov AS, Geyl KK, Tarasenko MV, Gureev MA, Boyarskiy VP. π-π Noncovalent Interaction Involving 1,2,4- and 1,3,4-Oxadiazole Systems: The Combined Experimental, Theoretical, and Database Study. Molecules 2021; 26:5672. [PMID: 34577142 PMCID: PMC8466036 DOI: 10.3390/molecules26185672] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
A series of N-pyridyl ureas bearing 1,2,4- (1a, 2a, and 3a) and 1,3,4-oxadiazole moiety (1b, 2b, 3b) was prepared and characterized by HRMS, 1H and 13C NMR spectroscopy, as well as X-ray diffraction. The inspection of the crystal structures of (1-3)a,b and the Hirshfeld surface analysis made possible the recognition of the (oxadiazole)···(pyridine) and (oxadiazole)···(oxadiazole) interactions. The presence of these interactions was confirmed theoretically by DFT calculations, including NCI analysis for experimentally determined crystal structures as well as QTAIM analysis for optimized equilibrium structures. The preformed database survey allowed the verification of additional examples of relevant (oxadiazole)···π interactions both in Cambridge Structural Database and in Protein Data Bank, including the cocrystal of commercial anti-HIV drug Raltegravir.
Collapse
Affiliation(s)
- Sergey V. Baykov
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., 199034 Saint Petersburg, Russia; (S.V.B.); (A.S.M.); (A.S.N.); (K.K.G.)
| | - Alexander S. Mikherdov
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., 199034 Saint Petersburg, Russia; (S.V.B.); (A.S.M.); (A.S.N.); (K.K.G.)
| | - Alexander S. Novikov
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., 199034 Saint Petersburg, Russia; (S.V.B.); (A.S.M.); (A.S.N.); (K.K.G.)
| | - Kirill K. Geyl
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., 199034 Saint Petersburg, Russia; (S.V.B.); (A.S.M.); (A.S.N.); (K.K.G.)
| | - Marina V. Tarasenko
- Pharmaceutical Technology Transfer Centre, Yaroslavl State Pedagogical University Named after K.D. Ushinsky, 108 Respublikanskaya St., 150000 Yaroslavl, Russia;
| | - Maxim A. Gureev
- Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Vadim P. Boyarskiy
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., 199034 Saint Petersburg, Russia; (S.V.B.); (A.S.M.); (A.S.N.); (K.K.G.)
| |
Collapse
|
28
|
Patil S, Bhandari S. A Review: Discovering 1,3,4-oxadiazole and chalcone nucleus for cytotoxicity/EGFR inhibitory anticancer activity. Mini Rev Med Chem 2021; 22:805-820. [PMID: 34477516 DOI: 10.2174/1389557521666210902160644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/19/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Cancer is reported to be one of the most life-threatening diseases. Major limitations of currently used anticancer agents are drug resistance, very small therapeutic index, and severe, multiple side effects. OBJECTIVE The current scenario necessitates developing new anticancer agents, acting on novel targets for effectively controlling cancer. The epidermal growth factor receptor is one such target, which is being explored for 1,3,4-oxadiazole and chalcone nuclei. METHOD Findings of different researchers working on these scaffolds have been reviewed and analyzed, and the outcomes were summarized. This review focuses on Structure-Activity Relationship studies (SARs) and computational studies of various 1,3,4-oxadiazole and chalcone hybrids/derivatives reported as cytotoxic/EGFR-TK inhibitory anticancer activity. RESULT AND CONCLUSION 1,3,4-oxadiazole and chalcone hybrids/derivatives with varied substitutions are found to be effective pharmacophores in obtaining potent anticancer activity. Having done a thorough literature survey, we conclude that this review will surely provide firm and better insights to the researchers to design and develop potent hybrids/derivatives that inhibit EGFR.
Collapse
Affiliation(s)
- Shital Patil
- All India Shri Shivaji Memorial Society's College of Pharmacy, Kennedy Road, Near RTO, Pune-411001, India
| | - Shashikant Bhandari
- All India Shri Shivaji Memorial Society's College of Pharmacy, Kennedy Road, Near RTO, Pune-411001, India
| |
Collapse
|
29
|
Alam MM, Nazreen S, Almalki ASA, Elhenawy AA, Alsenani NI, Elbehairi SEI, Malebari AM, Alfaifi MY, Alsharif MA, Alfaifi SYM. Naproxen Based 1,3,4-Oxadiazole Derivatives as EGFR Inhibitors: Design, Synthesis, Anticancer, and Computational Studies. Pharmaceuticals (Basel) 2021; 14:870. [PMID: 34577570 PMCID: PMC8469912 DOI: 10.3390/ph14090870] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
A library of novel naproxen based 1,3,4-oxadiazole derivatives (8-16 and 19-26) has been synthesized and screened for cytotoxicity as EGFR inhibitors. Among the synthesized hybrids, compound2-(4-((5-((S)-1-(2-methoxynaphthalen-6-yl)ethyl)-1,3,4-oxadiazol-2-ylthio)methyl)-1H-1,2,3-triazol-1-yl)phenol(15) was the most potent compound against MCF-7 and HepG2cancer cells with IC50 of 2.13 and 1.63 µg/mL, respectively, and was equipotent to doxorubicin (IC50 1.62 µg/mL) towards HepG2. Furthermore, compound 15 inhibited EGFR kinase with IC50 0.41 μM compared to standard drug Erlotinib (IC50 0.30 μM). The active compound induces a high percentage of necrosis towards MCF-7, HePG2 and HCT 116 cells. The docking studies, DFT and MEP also supported the biological data. These results demonstrated that these synthesized naproxen hybrids have EGFR inhibition effects and can be used as leads for cancer therapy.
Collapse
Affiliation(s)
- Mohammad Mahboob Alam
- Department of Chemistry, Faculty of Science, Albaha University, Albaha 65731, Saudi Arabia; (A.A.E.); (N.I.A.)
| | - Syed Nazreen
- Department of Chemistry, Faculty of Science, Albaha University, Albaha 65731, Saudi Arabia; (A.A.E.); (N.I.A.)
| | | | - Ahmed A. Elhenawy
- Department of Chemistry, Faculty of Science, Albaha University, Albaha 65731, Saudi Arabia; (A.A.E.); (N.I.A.)
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City 11884, Egypt
| | - Nawaf I. Alsenani
- Department of Chemistry, Faculty of Science, Albaha University, Albaha 65731, Saudi Arabia; (A.A.E.); (N.I.A.)
| | - Serag Eldin I. Elbehairi
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia; (S.E.I.E.); (M.Y.A.)
- Cell Culture Laboratory, Egyptian Organization for Biological Products and Vaccines, VACSERA Holding Company, Giza 22311, Egypt
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad Y. Alfaifi
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia; (S.E.I.E.); (M.Y.A.)
| | - Meshari A. Alsharif
- Chemistry Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21421, Saudi Arabia;
| | - Sulaiman Y. M. Alfaifi
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
30
|
Effect of pyrrolo[3,4-d]pyridazinone derivatives in neuroinflammation induced by preincubation with lipopolysaccharide or coculturing with microglia-like cells. Biomed Pharmacother 2021; 141:111878. [PMID: 34243096 DOI: 10.1016/j.biopha.2021.111878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 01/15/2023] Open
Abstract
Alzheimer's disease is one of the most serious disorders of the 21st century. There is still no effective therapy for this condition. The study investigated the potential regenerative effect of four pyrrolo[3,4-d]pyridazinone derivatives in cultures of SH-SY5Y neuron-like cells preincubated with lipopolysaccharide (LPS) or cocultured with microglia-like cells. In addition to the traditional investigation of the effect on viability, the level of free radicals and nitric oxide, the average length of neurites was also measured. Via in silico studies, the possibility of penetration of the blood-brain barrier (BBB) by the tested compounds was assessed. The administration of LPS to the culture of SH-SY5Y cells as well as coculturing with microglia-like cells had a significant negative effect on the results of all the assays performed. The treatment with the tested derivatives in most cases significantly reduced this negative effect. The obtained results suggest that the compound L2 may have a beneficial impact on neuronal damage caused by LPS or proinflammatory cytokines secreted by microglia-like cells. Importantly, tested compounds can pass through the BBB, which allows them to enter the brain.
Collapse
|
31
|
Santos RVC, Cunha EGC, de Mello GSV, Rizzo JÂ, de Oliveira JF, do Carmo Alves de Lima M, da Rocha Pitta I, da Rocha Pitta MG, de Melo Rêgo MJB. New Oxazolidines Inhibit the Secretion of IFN-γ and IL-17 by PBMCS from Moderate to Severe Asthmatic Patients. Med Chem 2021; 17:289-297. [PMID: 32914717 DOI: 10.2174/1573406416666200910151950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Moderate to severe asthma could be induced by diverse proinflammatory cytokines, as IL-17 and IFN-γ, which are also related to treatment resistance and airway hyperresponsiveness. Oxazolidines emerged as a novel approach for asthma treatment, since some chemical peculiarities were suggested by previous studies. OBJECTIVE The present study aimed to evaluate the IL-17A and IFN-γ modulatory effect of two new oxazolidine derivatives (LPSF/NB-12 and -13) on mononucleated cells of patients with moderate and severe asthma. METHODS The study first looked at potential targets for oxazolidine derivatives using SWISS-ADME. After the synthesis of the compounds, cytotoxicity and cytokine levels were analyzed. RESULTS We demonstrated that LPSF/NB-12 and -13 reduced IFN-γ and IL-17 production in peripheral blood mononucleated cells from asthmatic patients in a concentrated manner. Our in silico analysis showed the neurokinin-1 receptor as a common target for both compounds, which is responsible for diverse proinflammatory effects of moderate and severe asthma. CONCLUSION The work demonstrated a novel approach against asthma, which deserves further studies of its mechanisms of action.
Collapse
Affiliation(s)
- Renata Virgínia Cavalcanti Santos
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Eudes Gustavo Constantino Cunha
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Gabriela Souto Vieira de Mello
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - José Ângelo Rizzo
- Servico de Pneumologia, Hospital das Clinicas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Jamerson Ferreira de Oliveira
- Laboratorio de Quimica e Inovacao Terapeutica (LQIT), Departamento de Antibioticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Maria do Carmo Alves de Lima
- Laboratorio de Quimica e Inovacao Terapeutica (LQIT), Departamento de Antibioticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Ivan da Rocha Pitta
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Maira Galdino da Rocha Pitta
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
32
|
ElZahabi HSA, Nafie MS, Osman D, Elghazawy NH, Soliman DH, El-Helby AAH, Arafa RK. Design, synthesis and evaluation of new quinazolin-4-one derivatives as apoptotic enhancers and autophagy inhibitors with potent antitumor activity. Eur J Med Chem 2021; 222:113609. [PMID: 34119830 DOI: 10.1016/j.ejmech.2021.113609] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022]
Abstract
This work presents the design and synthesis of a series of new quinazolin-4-one derivatives, based on the established effectiveness of quinazoline-based small molecules as anticancer agents. Synthesized compounds were more potent against MCF-7 than A-549 with low to submicromolar IC50s. Compound 17 exhibited the best IC50 being equipotent with the positive control doxorubicin (IC50 = 0.06 μM) and better than 5-fluorouracil (IC50 = 2.13 μM). Compound 17 was further tested against MDA-MB-231 and MCF-10A and was found to be > 2 folds more cytotoxic on MCF-7. Significant apoptotic activity was elicited by 17 on MCF-7 where it increased apoptotic cell death along with induction of pre-G1 and G1-phase cell cycle arrest. Similarly, 17 was able to induce apoptosis in MD-MB-231 treated cells associated with a disruption of the cell cycle causing arrest at the pre-G1 and S phases. Investigation of gene expression in MCF-7 demonstrated an increased expression of the proapoptotic genes P53, PUMA, Bax, caspases 3, 8 and 9 and a decrease of the anti-apoptotic gene Bcl2. Also, 17 reduced autophagy giving way for apoptosis to induce cancer cells death. This latter observation was associated with downregulation of EGFR and its downstream effectors PI3K, AKT and mTor. As its biomolecular target, 17 also inhibited EGFR similar to erlotinib (IC50 = 0.072 and 0.087 μM, respectively). Additionally, in vivo testing in a mouse model of breast cancer affirmed the anti-tumor efficacy of 17. Finally, docking of 17 against EGFR ATP binding site demonstrated its ability to bind with EGFR resembling erlotinib.
Collapse
Affiliation(s)
- Heba S A ElZahabi
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Girls Branch, Cairo, Egypt
| | - Mohamed S Nafie
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| | - Dina Osman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, MSA University, Egypt
| | - Nehal H Elghazawy
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Dalia H Soliman
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Girls Branch, Cairo, Egypt
| | - Abdelghany Ali H El-Helby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Boys Branch, Cairo, Egypt
| | - Reem K Arafa
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
| |
Collapse
|
33
|
Zhang L, Xu L, Chen H, Zhang W, Xing C, Qu Z, Yu J, Zhuang C. Structure-based molecular hybridization design of Keap1-Nrf2 inhibitors as novel protective agents of acute lung injury. Eur J Med Chem 2021; 222:113599. [PMID: 34119834 DOI: 10.1016/j.ejmech.2021.113599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022]
Abstract
Blocking the Kelch-like epichlorohydrin-related protein 1 (Keap1)-nuclear factor-erythroid 2 related factor 2 (Nrf2) pathway represents as a promising strategy to reduce oxidative stress and related-inflammation, including acute lung injury (ALI). NXPZ-2, a naphthalensulfonamide derivative, was previously reported to effectively inhibit the Keap1-Nrf2 protein-protein interaction (PPI) by our group. In the present work, a series of novel isothiocyanate-containing naphthalensulfonamides with the thioether, sulfoxide and sulfone moieties were designed by a structure-based molecular hybridization strategy using NXPZ-2 and the Nrf2 activator sulforaphane. They possessed good Keap1-Nrf2 PPI inhibitory activity and low cytotoxicity. The molecular docking study was performed to further explain the different activity of the thioether-, sulfoxide- and sulfone-containing naphthalensulfonamides. Among these new derivatives, 2-((N-(4-((N-(2-amino-2-oxoethyl)-4-((3-isothiocyanatopropyl)sulfinyl)phenyl)sulfonamido) naphthalen-1-yl)-4-methoxyphenyl)sulfonamido)acetamide (SCN-16) showed a good KD2 value of 0.455 μM to disrupt the PPI. In an LPS-induced peritoneal macrophage cell model, this compound could cause a significant increase in the nuclear Nrf2 protein, decrease in the cytosolic Nrf2 protein, and further elevate the downstream protective enzymes HO-1 and NQO-1, which were better than the lead compound NXPZ-2 and sulforaphane. What's more, the production of ROS and NO and the expression of pro-inflammatory cytokine TNF-α were also suppressed. In the LPS-induced ALI model, SCN-16 could significantly reduce LPS-induced inflammations and alleviate lung injuries by triggering Nrf2 nuclear translocation. Collectively, our results suggested that SCN-16 could be a novel lead compound targeting Keap1-Nrf2 protective pathway for clinical treatment of ALI.
Collapse
Affiliation(s)
- Le Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Lijuan Xu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Haihu Chen
- Department of Intervention, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, FL, 32610, USA
| | - Zhuo Qu
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
34
|
Abdelgawad MA, Musa A, Almalki AH, Alzarea SI, Mostafa EM, Hegazy MM, Mostafa-Hedeab G, Ghoneim MM, Parambi DGT, Bakr RB, Al-Muaikel NS, Alanazi AS, Alharbi M, Ahmad W, Bukhari SNA, Al-Sanea MM. Novel Phenolic Compounds as Potential Dual EGFR and COX-2 Inhibitors: Design, Semisynthesis, in vitro Biological Evaluation and in silico Insights. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2325-2337. [PMID: 34103896 PMCID: PMC8178614 DOI: 10.2147/dddt.s310820] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022]
Abstract
Introduction Epidermal growth factor receptor (EGFR) inhibition is an imperative therapeutic approach targeting various types of cancer including colorectal, lung, breast, and pancreatic cancer types. Moreover, cyclooxygenase-2 (COX-2) is frequently overexpressed in different types of cancers and has a role in the promotion of malignancy, apoptosis inhibition, and metastasis of tumor cells. Combination therapy has been emerged to improve the therapeutic benefit against cancer and curb intrinsic and acquired resistance. Methods Three semi-synthetic series of compounds (C1-4, P1-4, and G1-4) were prepared and evaluated biologically as potential dual epidermal growth factor receptor (EGFR) and COX-2 inhibitors. The main phenolic constituents of Amaranthus spinosus L. (p-coumaric, caffeic and gallic) acids have been isolated and subsequently subjected to diazo coupling with various amines to get novel three chemical scaffolds with potential anticancer activities. Results Compounds C4 and G4 showed superior inhibitory activity against EGFR (IC50: 0.9 and 0.5 µM, respectively) and displayed good COX-2 inhibition (IC50: 4.35 and 2.47 µM, respectively). Moreover, the final compounds were further evaluated for their cytotoxic activity against human colon cancer (HT-29), pancreatic cancer (PaCa-2), human malignant melanoma (A375), lung cancer (H-460), and pancreatic ductal cancer (Panc-1) cell lines. Interestingly, compounds C4 and G4 exhibited the highest cytotoxic activity with average IC50 values of 1.5 µM and 2.8 µM against H-460 and Panc-1, respectively. The virtual docking study was conducted to gain proper understandings of the plausible-binding modes of target compounds within EGFR and COX-2 binding sites. Discussion The NMR of prepared compounds showed characteristic peaks that confirmed the structure of the target compounds. The synthesized benzoxazolyl scaffold containing compounds showed inhibitory activities for both COXs and EGFR which are consistent with the virtual docking study.
Collapse
Affiliation(s)
- Mohamed A Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka, 72341, Saudi Arabia
| | - Atiah H Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, 21944, Saudi Arabia.,Addiction and Neuroscience Research Unit, Taif University, Taif, 21944, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Ehab M Mostafa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka, 72341, Saudi Arabia
| | - Mostafa M Hegazy
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371, Egypt
| | - Gomaa Mostafa-Hedeab
- Department of Pharmacology, Medical College, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Mohammed M Ghoneim
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371, Egypt.,Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, 13713, Saudi Arabia
| | - Della G T Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Rania B Bakr
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Nayef S Al-Muaikel
- Department of Chemistry, College of Science, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Abdullah S Alanazi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Aljouf, Saudi Arabia.,Health Sciences Research Unit, Jouf University, Sakaka, Aljouf, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Waqas Ahmad
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Syed N A Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Mohammad M Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| |
Collapse
|
35
|
Febuxostat-based amides and some derived heterocycles targeting xanthine oxidase and COX inhibition. Synthesis, in vitro and in vivo biological evaluation, molecular modeling and in silico ADMET studies. Bioorg Chem 2021; 113:104948. [PMID: 34052736 DOI: 10.1016/j.bioorg.2021.104948] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022]
Abstract
Various febuxostat derivatives comprising carboxamide functionalities and different substituted heterocycles were synthesized and evaluated for their biological activities as xanthine oxidase (XO) and cyclooxygenase (COX) inhibitors. All the tested compounds exhibited variable in vitro XO inhibitory activities (IC50 values 0.009-0.077 µM), among which the analog 17 has emerged as the most potent derivative (IC50 0.009 µM), representing nearly 3-times the potency of febuxostat (IC50 0.026 µM). The same analogs were further investigated for their in vitro COX-1 and COX-2 inhibitory activity, where fifteen analogs demonstrated recognizable COX-2 inhibitory potential (IC50 values range 0.04 - 0.1 µM), when correlated with celecoxib (IC50 0.05 µM), together with appreciable selectivity indices. Compounds 5a, 14b, 17, 19c, 19e and 21b that showed significant in vitro XO and/ or COX inhibitory potentials were further investigated for their in vivo hypouricemic as well as anti-inflammatory activities. Interestingly, the in vivo results were concordant with the collected in vitro data. Docking of compounds 5a, 14b, 17, 19c, 19e and 21b with the active sites of XO and COX-2 isozymes demonstrated superior binding profile compared with the reported ligands (febuxostat and celecoxib, respectively). Their docking scores were reasonable and cohering to a great extent with their corresponding in vitro IC50 values. Moreover, in silico computation of the predicted pharmacokinetic and toxicity properties (ADMET), together with the ligand efficiency (LE) of the same six compounds suggesting their liability to act as new orally active drug candidates with a predicted high safety profile.
Collapse
|
36
|
Amin S, Alam MM, Akhter M, Najmi AK, Siddiqui N, Husain A, Shaquiquzzaman M. A review on synthetic procedures and applications of phosphorus oxychloride (POCl 3) in the last biennial period (2018–19). PHOSPHORUS SULFUR 2021. [DOI: 10.1080/10426507.2020.1831499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shaista Amin
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - M. Mumtaz Alam
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mymoona Akhter
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - A. K. Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nadeem Siddiqui
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Asif Husain
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - M. Shaquiquzzaman
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
37
|
Antiproliferative activity, enzymatic inhibition and apoptosis-promoting effects of benzoxazole-based hybrids on human breast cancer cells. Bioorg Chem 2021; 109:104752. [PMID: 33657444 DOI: 10.1016/j.bioorg.2021.104752] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/26/2021] [Accepted: 02/15/2021] [Indexed: 12/30/2022]
Abstract
New benzoxazole derivatives containing 1,3,4-oxadiazole, 1,2,4-triazole or triazolothiadiazine rings were synthesized and screened for their in vitro antiproliferative activities against MCF-7 and MDA-MB-231 breast cancer cell lines using MTT assay. Doxorubicin, cisplatin and 2-(4-aminophenyl)benzothiazole (CJM 126) were used as references. The most active compounds 7a, 8d, 8e and 10c were screened for their antiproliferative activities against MCF-10A normal breast cells where compounds 8e and 7a were the most selective towards MCF-7 and MDA-MB-231 cell lines, respectively compared to CJM 126. In vitro enzymatic inhibition assays of epidermal growth factor receptor (EGFR) and aromatase (ARO) enzymes were performed. Compound 7a showed inhibition of EGFR comparable to that of erlotinib while compound 8e exhibited nearly half the inhibitory activity of erlotinib towards EGFR and was more potent inhibitor of ARO than letrozole. Caspase-9 activation assay, cell cycle analysis and Annexin-V/ Propidium iodide assay performed for compounds 7a, 8d, 8e and 10c demonstrated over expression of caspase-9 protein level, pre G1 apoptosis and high annexin V binding affinity. Therefore, these compounds are considered as potent apoptosis-promoting agents. The predicted docking studies and in silico chemo-informatic properties of compounds 7a and 8e were appropriate. Compounds 7a and 8e are promising anti-breast cancer agents exhibiting potent apoptosis-promoting properties.
Collapse
|
38
|
LncRNA TINCR favors tumorigenesis via STAT3-TINCR-EGFR-feedback loop by recruiting DNMT1 and acting as a competing endogenous RNA in human breast cancer. Cell Death Dis 2021; 12:83. [PMID: 33446634 PMCID: PMC7809450 DOI: 10.1038/s41419-020-03188-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022]
Abstract
The long noncoding RNA (lncRNA) TINCR has recently been found to be associated with the progression of human malignancies, but the molecular mechanism of TINCR action remains elusive, particularly in breast cancer. The oncogenic role of TINCR was examined in vitro and in vivo in breast cancer. Next, the interaction between TINCR, DNMT1, and miR-503-5p methylation was explored. Moreover, the mechanism by which TINCR enhances EGFR expression and downstream signaling via an RNA–RNA interaction was comprehensively investigated. Furthermore, upstream transcriptional regulation of TINCR expression by STAT3 was examined by performing chromatin immunoprecipitation. Finally, feedback signaling in the STAT3–TINCR–EGFR downstream cascade was also investigated. TINCR is upregulated in human breast cancer tissues, and TINCR knockdown suppresses tumorigenesis in vitro and in vivo. Mechanistically, TINCR recruits DNMT1 to the miR-503-5p locus promoter, which increases the methylation and suppresses the transcriptional expression of miR-503-5p. Furthermore, TINCR also functions as a competing endogenous RNA to upregulate EGFR expression by sponging miR-503-5p. In addition, TINCR stimulates JAK2–STAT3 signaling downstream from EGFR, and STAT3 reciprocally enhances the transcriptional expression of TINCR. Our findings broaden the current understanding of the diverse manners in which TINCR functions in cancer biology. The newly identified STAT3–TINCR–EGFR-feedback loop could serve as a potential therapeutic target for human cancer.
Collapse
|
39
|
Novel 1,3,4-Oxadiazole Derivatives of Pyrrolo[3,4- d]pyridazinone Exert Antinociceptive Activity in the Tail-Flick and Formalin Test in Rodents and Reveal Reduced Gastrotoxicity. Int J Mol Sci 2020; 21:ijms21249685. [PMID: 33353118 PMCID: PMC7766312 DOI: 10.3390/ijms21249685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the availability of the current drug arsenal for pain management, there is still a clinical need to identify new, more effective, and safer analgesics. Based on our earlier study, newly synthesized 1,3,4-oxadiazole derivatives of pyrrolo[3,4-d]pyridazinone, especially 10b and 13b, seem to be promising as potential analgesics. The current study was designed to investigate whether novel derivatives attenuate nociceptive response in animals subjected to thermal or chemical noxious stimulus, and to compare this effect to reference drugs. The antinociceptive effect of novel compounds was studied using the tail-flick and formalin test. Pretreatment with novel compounds at all studied doses increased the latency time in the tail-flick test and decreased the licking time during the early phase of the formalin test. New derivatives given at the medium and high doses also reduced the late phase of the formalin test. The achieved results indicate that new derivatives dose-dependently attenuate nociceptive response in both models of pain and exert a lack of gastrotoxicity. Both studied compounds act more efficiently than indomethacin, but not morphine. Compound 13b at the high dose exerts the greatest antinociceptive effect. It may be due to the reduction of nociceptor sensitization via prostaglandin E2 and myeloperoxidase levels decrease.
Collapse
|
40
|
Szczukowski Ł, Krzyżak E, Zborowska A, Zając P, Potyrak K, Peregrym K, Wiatrak B, Marciniak A, Świątek P. Design, Synthesis and Comprehensive Investigations of Pyrrolo[3,4- d]pyridazinone-Based 1,3,4-Oxadiazole as New Class of Selective COX-2 Inhibitors. Int J Mol Sci 2020; 21:E9623. [PMID: 33348757 PMCID: PMC7766220 DOI: 10.3390/ijms21249623] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
The long-term use of Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) in treatment of different chronic inflammatory disorders is strongly restricted by their serious gastrointestinal adverse effects. Therefore, there is still an urgent need to search for new, safe, and efficient anti-inflammatory agents. Previously, we have reported the Mannich base-type derivatives of pyrrolo[3,4-d]pyridazinone which strongly inhibit cyclooxygenase, have better affinity to COX-2 isoenzyme and exert promising anti-oxidant activity. These findings encouraged us to perform further optimization of that structure. Herein, we present the design, synthesis, molecular docking, spectroscopic, and biological studies of novel pyrrolo[3,4-d]pyridazinone derivatives bearing 4-aryl-1-(1-oxoethyl)piperazine pharmacophore 5a,b-6a,b. The new compounds were obtained via convenient, efficient, one-pot synthesis. According to in vitro evaluations, novel molecules exert no cytotoxicity and act as selective COX-2 inhibitors. These findings stay in good correlation with molecular modeling results, which additionally showed that investigated compounds take a position in the active site of COX-2 very similar to Meloxicam. Moreover, all derivatives reduce the increased level of reactive oxygen and nitrogen species and prevent DNA strand breaks caused by oxidative stress. Finally, performed spectroscopic and molecular docking studies demonstrated that new compound interactions with bovine serum albumin (BSA) are moderate, formation of complexes is in one-to-one ratio, and binding site II (subdomain IIIA) is favorable.
Collapse
Affiliation(s)
- Łukasz Szczukowski
- Department of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| | - Edward Krzyżak
- Department of Inorganic Chemistry, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland; (E.K.); (A.M.)
| | - Adrianna Zborowska
- Student Scientific Club of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.Z.); (P.Z.); (K.P.); (K.P.)
| | - Patrycja Zając
- Student Scientific Club of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.Z.); (P.Z.); (K.P.); (K.P.)
| | - Katarzyna Potyrak
- Student Scientific Club of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.Z.); (P.Z.); (K.P.); (K.P.)
| | - Krzysztof Peregrym
- Student Scientific Club of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.Z.); (P.Z.); (K.P.); (K.P.)
| | - Benita Wiatrak
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland
| | - Aleksandra Marciniak
- Department of Inorganic Chemistry, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland; (E.K.); (A.M.)
| | - Piotr Świątek
- Department of Medicinal Chemistry, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| |
Collapse
|
41
|
In Vitro and In Silico Evaluation of Anticancer Activity of New Indole-Based 1,3,4-Oxadiazoles as EGFR and COX-2 Inhibitors. Molecules 2020; 25:molecules25215190. [PMID: 33171861 PMCID: PMC7664637 DOI: 10.3390/molecules25215190] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and cyclooxygenase-2 (COX-2) are crucial targetable enzymes in cancer management. Therefore, herein, new 2-[(5-((1H-indol-3-yl)methyl)-1,3,4-oxadiazol-2-yl)thio]-N-(thiazol/benzothiazol-2-yl)acetamides (2a-i) were designed and synthesized as EGFR and COX-2 inhibitors. The cytotoxic effects of compounds 2a-i on HCT116 human colorectal carcinoma, A549 human lung adenocarcinoma, and A375 human melanoma cell lines were determined using MTT assay. 2-[(5-((1H-Indol-3-yl)methyl)-1,3,4-oxadiazol-2-yl)thio]-N-(6-ethoxybenzothiazol-2-yl)acetamide (2e) exhibited the most significant anticancer activity against HCT116, A549, and A375 cell lines with IC50 values of 6.43 ± 0.72 μM, 9.62 ± 1.14 μM, and 8.07 ± 1.36 μM, respectively, when compared with erlotinib (IC50 = 17.86 ± 3.22 μM, 19.41 ± 2.38 μM, and 23.81 ± 4.17 μM, respectively). Further mechanistic assays demonstrated that compound 2e enhanced apoptosis (28.35%) in HCT116 cells more significantly than erlotinib (7.42%) and caused notable EGFR inhibition with an IC50 value of 2.80 ± 0.52 μM when compared with erlotinib (IC50 = 0.04 ± 0.01 μM). However, compound 2e did not cause any significant COX-2 inhibition, indicating that this compound showed COX-independent anticancer activity. The molecular docking study of compound 2e emphasized that the benzothiazole ring of this compound occupied the allosteric pocket in the EGFR active site. In conclusion, compound 2e is a promising EGFR inhibitor that warrants further clinical investigations.
Collapse
|
42
|
|
43
|
Design, synthesis, biological evaluation and in silico studies of novel pyrrolo[3,4-d]pyridazinone derivatives with promising anti-inflammatory and antioxidant activity. Bioorg Chem 2020; 102:104035. [DOI: 10.1016/j.bioorg.2020.104035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023]
|
44
|
Xu C, Han Y, Xu S, Wang R, Yue M, Tian Y, Li X, Zhao Y, Gong P. Design, synthesis and biological evaluation of new Axl kinase inhibitors containing 1,3,4-oxadiazole acetamide moiety as novel linker. Eur J Med Chem 2020; 186:111867. [PMID: 31757525 DOI: 10.1016/j.ejmech.2019.111867] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022]
Abstract
Using the principle of bioisosteric replacement, we present a structure-based design approach to obtain new Axl kinase inhibitors with significant activity at the kinase and cellular levels. Through a stepwise structure-activity relationships exploration, a series of 6,7-disubstituted quinoline derivatives, which contain 1,3,4-oxadiazol acetamide moiety as novel Linker, were ultimately synthesized with Axl as the primary target. Most of them exhibited moderate to excellent activity, with IC50 values ranging from 0.032 to 1.54 μM against the tested cell lines. Among them, the most promising compound 47e, as an Axl kinase inhibitor (IC50 = 10 nM), shows remarkable cytotoxicity against A549, HT-29, PC-3, MCF-7, H1975 and MDA-MB-231 cell lines. More importantly, 47e also shows a significant inhibitory effect on EGFR-TKI resistant NSCLC cell lines H1975/gefitinib. Meanwhile, this study provides a novel type of linker for Axl kinase inhibitors, namely 1,3,4-oxadiazol acetamide moiety, which is out of the scope of the "5- atoms role ".
Collapse
Affiliation(s)
- Congjun Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Yufei Han
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China
| | - Sicong Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China
| | - Ruxin Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China
| | - Ming Yue
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China
| | - Yu Tian
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China
| | - Xiaofan Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China
| | - Yanfang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China.
| | - Ping Gong
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, PR China.
| |
Collapse
|
45
|
Kharkar PS. Computational Approaches for the Design of (Mutant-)Selective Tyrosine Kinase Inhibitors: State-of-the-Art and Future Prospects. Curr Top Med Chem 2020; 20:1564-1575. [PMID: 32357816 DOI: 10.2174/1568026620666200502005853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/10/2020] [Accepted: 03/26/2020] [Indexed: 02/08/2023]
Abstract
Kinases remain one of the major attractive therapeutic targets for a large number of indications such as cancer, rheumatoid arthritis, cardiac failure and many others. Design and development of kinase inhibitors (ATP-competitive, allosteric or covalent) is a clinically validated and successful strategy in the pharmaceutical industry. The perks come with limitations, particularly the development of resistance to highly potent and selective inhibitors. When this happens, the cycle needs to be repeated, i.e., the design and development of kinase inhibitors active against the mutated forms. The complexity of tumor milieu makes it awfully difficult for these molecularly-targeted therapies to work. Every year newer and better versions of these agents are introduced in the clinic. Several computational approaches such as structure-, ligand-based or hybrid ones continue to live up to their potential in discovering novel kinase inhibitors. New schools of thought in this area continue to emerge, e.g., development of dual-target kinase inhibitors. But there are fundamental issues with this approach. It is indeed difficult to selectively optimize binding at two entirely different or related kinases. In addition to the conventional strategies, modern technologies (machine learning, deep learning, artificial intelligence, etc.) started yielding the results and building success stories. Computational tools invariably played a critical role in catalysing the phenomenal progress in kinase drug discovery field. The present review summarized the progress in utilizing computational methods and tools for discovering (mutant-)selective tyrosine kinase inhibitor drugs in the last three years (2017-2019). Representative investigations have been discussed, while others are merely listed. The author believes that the enthusiastic reader will be inspired to dig out the cited literature extensively to appreciate the progress made so far and the future prospects of the field.
Collapse
Affiliation(s)
- Prashant S Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai 400 019, India
| |
Collapse
|