1
|
Tambekar A, Guhe V, Singh S. The cGAS-STING mediated crosstalk between innate immunity and autophagy in leishmaniasis using mathematical modeling: Uncovering new therapeutic avenues. Arch Biochem Biophys 2024; 762:110201. [PMID: 39486567 DOI: 10.1016/j.abb.2024.110201] [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: 07/15/2024] [Revised: 09/11/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
The present paper deals with the investigation into the cGAS-STING pathway, focusing on the signaling of interferons through mathematical modeling and identifying a significant positive feedback loop regulated by STING for activation of type 1 interferons (IFN-1). Cyclic GMP-AMP synthase (cGAS) is responsible for detecting cytosolic DNA and initiating the STING (stimulator of interferon genes) pathway, which in turn causes the synthesis of pro-inflammatory cytokines and type I interferons. In addition to being crucial for pathogen identification, this route interacts with autophagy, a cellular mechanism that is necessary for immunological homeostasis and pathogen removal. In the context of Leishmania infection, the cGAS-STING signaling axis has come to light as a critical mediator of the crosstalk between innate immunity and autophagy. Further, the protein-protein interaction studies underscored the significance of two distinct domains in mediating interactions with IRF3 and LC3. Importantly, our findings suggest the possibility of manipulating STING concomitantly to regulate IRF3 and LC3 independently. This study remarkably advances our understanding of STING's multifaceted roles, particularly in regulating IFN-1 and autophagy, highlighting its pivotal role as a cross-talk point in leishmaniasis.
Collapse
Affiliation(s)
- Anil Tambekar
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India
| | - Vrushali Guhe
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India
| | - Shailza Singh
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India.
| |
Collapse
|
2
|
Triantopoulou S, Roupa I, Shegani A, Pirmettis NN, Terzoudi GI, Chiotellis A, Tolia M, Damilakis J, Pirmettis I, Paravatou-Petsota M. Synthesis and Biological Evaluation of Novel Cationic Rhenium and Technetium-99m Complexes Bearing Quinazoline Derivative for Epidermal Growth Factor Receptor Targeting. Pharmaceutics 2024; 16:1213. [PMID: 39339249 PMCID: PMC11434983 DOI: 10.3390/pharmaceutics16091213] [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: 07/25/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Epidermal growth factor receptor (EGFR) plays a vital role in cell proliferation and survival, with its overexpression linked to various malignancies, including non-small cell lung cancer (NSCLC). Although EGFR tyrosine kinase inhibitors (TKIs) are a key therapeutic strategy, acquired resistance and relapse remain challenges. This study aimed to synthesize and evaluate novel rhenium-based complexes incorporating EGFR TKIs to enhance anticancer efficacy, particularly in radiosensitization. Methods: We synthesized a rhenium tricarbonyl complex (Complex 2) and its 99mTc analog (Complex 2') by incorporating triphenylphosphine instead of bromine as the monodentate ligand and PF6- as the counter-ion, resulting in a positively charged compound that forms cationic structures. Cytotoxicity and EGFR inhibition were evaluated in A431 cells overexpressing EGFR using MTT assays, Western blotting, and flow cytometry. Radiosensitization was tested through MTT and clonogenic assays. The 99mTc complex's radiochemical yield, stability, and lipophilicity were also assessed. Results: Complex 2 exhibited significant cytotoxicity with an IC50 of 2.6 μM and EGFR phosphorylation inhibition with an IC50 of 130.6 nM. Both complex 1 and 2 induced G0/G1 cell cycle arrest, with Complex 2 causing apoptosis. Radiosensitization was observed at doses above 2 Gy. Complex 2' demonstrated high stability and favorable lipophilicity (LogD7.4 3.2), showing 12% cellular uptake after 30 min. Conclusions: Complexes 2 and 2' show promise as dual-function anticancer agents, offering EGFR inhibition, apoptosis induction, and radiosensitization. Their potential as radiopharmaceuticals warrants further in-depth investigation in preclinical models.
Collapse
Affiliation(s)
- Sotiria Triantopoulou
- Department of Medical Physics, School of Medicine, University of Crete, P.O. Box 2208, 71003 Heraklion, Greece; (S.T.); (J.D.)
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Ioanna Roupa
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Antonio Shegani
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Nektarios N. Pirmettis
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Georgia I. Terzoudi
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Aristeidis Chiotellis
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Maria Tolia
- Department of Radiation Oncology, University Hospital of Iraklion, 71110 Iraklion, Greece;
| | - John Damilakis
- Department of Medical Physics, School of Medicine, University of Crete, P.O. Box 2208, 71003 Heraklion, Greece; (S.T.); (J.D.)
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| | - Maria Paravatou-Petsota
- Institute of Nuclear and Radiological Sciences and Technology, Energy & Safety, NCSR “Demokritos”, P.O. Box 60037, 15310 Athens, Greece; (I.R.); (A.S.); (N.N.P.); (G.I.T.); (A.C.)
| |
Collapse
|
3
|
Al-Sheikh A, Jaber MA, Khalaf H, AlKhawaja N, Abuarqoub D. Synthesis and biological evaluation of novel 2-morpholino-4-anilinoquinoline derivatives as antitumor agents against HepG2 cell line. RSC Adv 2024; 14:3304-3313. [PMID: 38249681 PMCID: PMC10798140 DOI: 10.1039/d3ra07495a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Cancer is a life-threatening illness all over the world, and developing anticancer treatments with high efficacy and low side effects remains a challenge. The quinoline ring structure has long been recognized as a flexible nucleus in the design and synthesis of physiologically active chemicals. In this study, five new 2-morpholino-4-anilinoquinoline compounds were synthesized and their biological anticancer potential against the HepG2 cell line was assessed. The compounds produced demonstrated varying responses against HepG2 cells, with compounds 3c, 3d, and 3e exhibiting the highest activity, with IC50 values of 11.42, 8.50, and 12.76 μM, respectively. It is a critical requirement that anticancer medications are able to selectively decrease cancer growth while not causing damage to normal cells. Compound 3e exhibited increased activity while maintaining adequate selectivity. It was also the most effective chemical against cell migration and adhesion, which could play an important role in drug resistance and cell metastasis. In total, the findings revealed good possibilities for anticancer therapy, suggesting a target for future development of anticancer medication.
Collapse
Affiliation(s)
- Ahmed Al-Sheikh
- Department of Pharmaceutical Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
| | - Malak A Jaber
- Department of Pharmaceutical Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
| | - Hana'a Khalaf
- Department of Clinical Nutrition and Diets, Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
| | - Nour AlKhawaja
- Pharmaceutical Studies Center, Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
| | - Duaa Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
- Cell Therapy Center, University of Jordan Amman 11942 Jordan
| |
Collapse
|
4
|
Anandu KR, Jayan AP, Aneesh TP, Saiprabha VN. Pyrimidine derivatives as EGFR tyrosine kinase inhibitors in NSCLC: - A comprehensive review. Chem Biol Drug Des 2022; 100:599-621. [PMID: 35883248 DOI: 10.1111/cbdd.14124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/11/2022] [Accepted: 07/17/2022] [Indexed: 11/30/2022]
Abstract
EGFR positive NSCLC due to primary mutation (EGFR DEL19 & L858R) has been recognized as a crucial mediator of tumor progression. This led to the development and approval of EGFR tyrosine kinase inhibitors which addresses EGFR mediated NSCLC but fail to show potency after initial months of therapy due to acquired resistance (EGFR T790M, EGFR C797S). Extensive research allowed identification of drugs for EGFR positive NSCLC, wherein the majority of compounds have a pyrimidine substructure offering marked therapeutic benefits compared to chemotherapy. This current review outlines the diverse pyrimidine derivatives with amino-linked and fused pyrimidine scaffolds such as furo-pyrimidine, pyrimido-pyrimidine, thieno-pyrimidine, highlighting pyrimidine EGFR TK inhibitors reported in research emphasizing structural aspects, design approaches, inhibition potential. selectivity profile towards mutant EGFR conveyed through biological evaluation studies. Furthermore, mentioning the in-silico interaction profile of synthesized compounds for evaluating the binding affinity with key amino acids. The epilogue of review focuses on the recent research that drives forward to aid in the discovery and development of substituted amino and fused scaffolds of pyrimidine that can counteract the mutations and effectively manage EGFR positive NSCLC.
Collapse
Affiliation(s)
- K R Anandu
- Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Ajay P Jayan
- Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - T P Aneesh
- Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - V N Saiprabha
- Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| |
Collapse
|
5
|
Haider K, Das S, Joseph A, Yar MS. An appraisal of anticancer activity with structure-activity relationship of quinazoline and quinazolinone analogues through EGFR and VEGFR inhibition: A review. Drug Dev Res 2022; 83:859-890. [PMID: 35297084 DOI: 10.1002/ddr.21925] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/29/2022] [Accepted: 02/06/2022] [Indexed: 12/28/2022]
Abstract
Cancer is one of the leading causes of death. Globally a huge number of deaths and new incidences are reported annually. Heterocyclic compounds have been proved to be very effective in the treatment of different types of cancer. Among different heterocyclic scaffolds, quinazoline and quinazolinone core were found versatile and interesting with many biological activities. In the discovery of novel anticancer agents, the Quinazoline core is very effective. The FDA has approved more than 20 drugs as an anticancer bearing quinazoline or quinazolinone core in the last two decades. One prime example is Dacomitinib, which was newly approved for non-small-cell lung carcinoma treatment in 2018. These drugs work by different pathways to prevent the spread of cancer cell progression, including inhibition of different kinases, tubulin, kinesin spindle protein, and so forth. This review presented recent developments of quinazoline/quinazolinone scaffold bearing derivatives as anticancer agents acting as epidermal growth factor receptor (EGFR) vascular endothelial growth factor receptor (VEGFR), and dual EGFR/VEGFR inhibitors.
Collapse
Affiliation(s)
- Kashif Haider
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,Centre for Excellence for Biomaterials Engineering, Faculty of Applied Sciences, AIMST University, Malaysia
| |
Collapse
|
6
|
Hricovíniová J, Hricovíniová Z, Kozics K. Antioxidant, Cytotoxic, Genotoxic, and DNA-Protective Potential of 2,3-Substituted Quinazolinones: Structure-Activity Relationship Study. Int J Mol Sci 2021; 22:E610. [PMID: 33435390 PMCID: PMC7828088 DOI: 10.3390/ijms22020610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 11/30/2022] Open
Abstract
The evaluation of antioxidant compounds that counteract the mutagenic effects caused by the direct action of reactive oxygen species on DNA molecule is of considerable interest. Therefore, a series of 2,3-substituted quinazolinone derivatives (Q1-Q8) were investigated by different assays, and the relationship between their biological properties and chemical structure was examined. Genotoxicity and the potential DNA-protective effects of Q1-Q8 were evaluated by comet assay and DNA topology assay. Antioxidant activity was examined by DPPH-radical-scavenging, reducing-power, and total antioxidant status (TAS) assays. The cytotoxic effect of compounds was assessed in human renal epithelial cells (TH-1) and renal carcinoma cells (Caki-1) by MTT assay. Analysis of the structure-activity relationship disclosed significant differences in the activity depending on the substitution pattern. Derivatives Q5-Q8, bearing electron-donating moieties, were the most potent members of this series. Compounds were not genotoxic and considerably decreased the levels of DNA lesions induced by oxidants (H2O2, Fe2+ ions). Furthermore, compounds exhibited higher cytotoxicity in Caki-1 compared to that in TH-1 cells. Substantial antioxidant effect and DNA-protectivity along with the absence of genotoxicity suggested that the studied quinazolinones might represent potential model structures for the development of pharmacologically active agents.
Collapse
Affiliation(s)
- Jana Hricovíniová
- Cancer Research Institute BMC, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia;
| | - Zuzana Hricovíniová
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia;
| | - Katarína Kozics
- Cancer Research Institute BMC, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia;
| |
Collapse
|
7
|
Liu YM, Li Y, Liu RF, Xiao J, Zhou BN, Zhang QZ, Song JX. Synthesis, characterization and preliminary biological evaluation of chrysin amino acid derivatives that induce apoptosis and EGFR downregulation. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2021; 23:39-54. [PMID: 31833411 DOI: 10.1080/10286020.2019.1702028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 06/10/2023]
Abstract
Chrysin amino acid derivatives were synthesized to evaluate for their antiproliferative activities. Among them, N-(7-((5-hydroxy-4-oxo-2-phenyl-4H-chromen-7-yl)oxy)valeryl)-L-leucine (8c) displayed the most remarkable inhibitory activities against MCF-7 cells with IC50 values of 16.6 μM. Preliminary mechanistic studies showed that 8c could inhibit the colony formation and migration of MCF-7 cells. Flow cytometry analysis demonstrated that 8c mediated cell apoptosis and the prolongation of cell cycle progression in G1/S-phase against MCF-7 cells. Besides, 8c displayed the moderate inhibition against EGFR. Western blot assay suggested that 8c significantly inhibited EGFR phosphorylation. Molecular docking showed that 8c can bind the EGFR kinase well.
Collapse
Affiliation(s)
- Yun-Mei Liu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), Key Laboratory of the Assembly and Application of Organic Functional molecules of Hunan Province, Hunan Normal University, Changsha 410081, China
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Yang Li
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Rong-Fang Liu
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Jie Xiao
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Bin-Ning Zhou
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Qi-Zhi Zhang
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Jian-Xin Song
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), Key Laboratory of the Assembly and Application of Organic Functional molecules of Hunan Province, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
8
|
Li Y, Xiao J, Zhang Q, Yu W, Liu M, Guo Y, He J, Liu Y. The association between anti-tumor potency and structure-activity of protein-kinases inhibitors based on quinazoline molecular skeleton. Bioorg Med Chem 2019; 27:568-577. [DOI: 10.1016/j.bmc.2018.12.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 02/03/2023]
|
9
|
Liu Z, Wang L, Feng M, Yi Y, Zhang W, Liu W, Li L, Liu Z, Li Y, Ma X. New acrylamide-substituted quinazoline derivatives with enhanced potency for the treatment of EGFR T790M-mutant non-small-cell lung cancers. Bioorg Chem 2018; 77:593-599. [DOI: 10.1016/j.bioorg.2018.01.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 01/21/2018] [Accepted: 01/27/2018] [Indexed: 11/25/2022]
|
10
|
Wang L, Zhao J, Yao Y, Wang C, Zhang J, Shu X, Sun X, Li Y, Liu K, Yuan H, Ma X. Covalent binding design strategy: A prospective method for discovery of potent targeted anticancer agents. Eur J Med Chem 2017; 142:493-505. [DOI: 10.1016/j.ejmech.2017.09.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 12/16/2022]
|
11
|
Ghorab MM, Alsaid MS, Soliman AM, Al-Mishari AA. Benzo[g]quinazolin-based scaffold derivatives as dual EGFR/HER2 inhibitors. J Enzyme Inhib Med Chem 2017; 33:67-73. [PMID: 29098904 PMCID: PMC6010119 DOI: 10.1080/14756366.2017.1389922] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Targeting EGFR has proven to be beneficial in the treatment of several types of solid tumours. So, a series of novel 2-(4-oxo-3-(4-sulfamoylphenyl)-3,4-dihydrobenzo[g]quinazolin-2-ylthio)-N-substituted acetamide 5–19 were synthesised from the starting material 4-(2-mercapto-4-oxobenzo[g]quinazolin-3(4H)-yl) benzenesulfonamide 4, to be evaluated as dual EGFR/HER2 inhibitors. The target compounds 5–19, were screened for their cytotoxic activity against A549 lung cancer cell line. The percentage inhibition of EGFR enzyme was measured and compared with erlotinib as the reference drug. Compounds 6, 8, 10, and 16 showed excellent EGFR inhibitory activity and were further selected for screening as dual EGFR/HER2 inhibitors. The four selected compounds showed IC50 ranging from 0.009 to 0.026 µM for EGFR and 0.021 to 0.069 µM for the HER2 enzyme. Compound 8 was found to be the most potent in this study with IC50 0.009 and 0.021 µM for EGFR and HER2, respectively.
Collapse
Affiliation(s)
- Mostafa M Ghorab
- a Department of Pharmacognosy, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia.,b Department of Drug Radiation Research , National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority , Cairo , Egypt
| | - Mansour S Alsaid
- a Department of Pharmacognosy, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Aiten M Soliman
- b Department of Drug Radiation Research , National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority , Cairo , Egypt
| | - Abdullah A Al-Mishari
- c Medicinal, Aromatic and Poisonous Plants Research Center (MAPPRC), College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|