1
|
Fadaly WAA, Nemr MTM, Abd El-Hameed AM, Mohamed FEA, Zidan TH. Design and Synthesis of New pyrazole Hybrids Linked to Oxime and Nitrate Moieties as COX-2, EGFR L858R/T790M Inhibitors and Nitric Oxide Donors with dual Anti-inflammatory/Anti-proliferative Activities. Bioorg Chem 2025; 161:108563. [PMID: 40349531 DOI: 10.1016/j.bioorg.2025.108563] [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: 01/08/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Two new series of pyrazole derivatives 14a-l and 17a-c with oxime/nitrate moieties as EGFRWT, mutant (EGFRL858R/T790M) and COX-2 inhibitors were synthesized and evaluated for anti-proliferative and anti-inflammatory activities. Compounds 14c, 14e, 14 g, 14i-l, 17b and 17c exhibited COX-2 selectivity in the range of (S.I. = 17-42) when compared to celecoxib (S.I. = 20.43). Concerning anti-neoplastic activity, screening was carried out against 60 human cancer cell lines by (NCI); Nine compounds (14c, 14e, 14 g, 14i-l, 17b and 17c) showed excellent inhibitory activity against all cancer cell lines especially non-small cell lung cancer (NSCLC). Further cytotoxicity testing of compounds 14c, 14e, 14 g, 14i-l, 17b and 17c was conducted on established EGFRT790M/L858R-resistant NSCLC (H1975), all tested compounds except 14 l exhibited potent activity (IC50 = 3.02-27.32 μM) which is higher than that of osimertinib (IC50 = 37.29 μM). It was noted that compound 17c, showed cell cycle arrest at G0/G1 phase of NSCLC (H1975) cells. In addition, compounds 14c, 14e, 14 g, 14i-l, 17b and 17c induced improved selective inhibitory activity against double mutant EGFRL858R/T790M tyrosine kinases with IC50 in the range of (0.031-0.076 μM, with selectivity index range S.I. of 2.5-14.58) which was comparable to that of osimertinib (IC50 = 0.037 μM, with S.I. of 1.89). The most potent anti-cancer compounds 14c, 14e, 14 g, 14i-l, 17b and 17c released NO in a slow rate of (1.45-3.37 %). Finally, applying covalent docking, we identified the covalent binding of 14 g, 14 k, and 17c with Cys797, providing insights into their potential as irreversible inhibitors targeting EGFRL858R/T790M protein.
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini Street, 11562 Cairo, Egypt.
| | - Abeer M Abd El-Hameed
- Chemistry Department, Faculty of Science, Taibah University, P.O. BOX 30002, Al-Madinah, Al-Munawarah 14177, Saudi Arabia
| | - Fatma E A Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Taha H Zidan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
2
|
Ding S, Wu Y, Li B, Wang X, Ding H, Zhan Z, Shen J, Qi R, Gao Z, Yao K, Su R, Zheng H, Tang Z, Liu J, Chen Y. Design, synthesis and biological evaluation of imine-containing inhibitors against the triple-mutant EGFR kinases based on the scaffold of osimertinib. Bioorg Chem 2025; 160:108474. [PMID: 40267777 DOI: 10.1016/j.bioorg.2025.108474] [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: 02/13/2025] [Revised: 04/06/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
A series of EGFR inhibitors were designed and synthesized based on the scaffold of osimertinib, the inhibitory activities against the L858R/T790M/C797S mutant EGFR kinase of which were subsequently evaluated. Compounds with the imine fragments showed the highest kinase inhibitory activity and were proved to be reversible inhibitors, which were represented by the compound DD-8 (IC50 = 0.87 nM). Kinase selectivity assay showed the compounds with imine fragments were preferred to inhibit the triple-mutant EGFR kinases rather than other subtypes. In the proliferation inhibition assay against the BaF3-EGFR(L858R/T790M/C797S) cell line, compound DD-8 also showed strong inhibitory activity (IC50 = 1.11 μM), and the inhibition rate of which reached 98.8 % at the concentration of 2 μM. To further elucidate the antitumor mechanism of the compound DD-8, a comprehensive series of experiments were conducted, including apoptosis induction assay, cell cycle arrest assay, western blot assay, cell migration inhibition assay, liver microsomal stability experiment, and molecular docking analysis.
Collapse
Affiliation(s)
- Shi Ding
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; API Engineering Technology Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Yuanyu Wu
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Bin Li
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Xin Wang
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Haotian Ding
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Zhenzhen Zhan
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Jiwei Shen
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; API Engineering Technology Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Rui Qi
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Ziye Gao
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Kai Yao
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Riya Su
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Hanxue Zheng
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Zhongyu Tang
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China
| | - Ju Liu
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; API Engineering Technology Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China.
| | - Ye Chen
- College of Pharmacy of Liaoning University, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; API Engineering Technology Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, 66 Chongshan Road, Huanggu District, Shenyang 110036, PR China.
| |
Collapse
|
3
|
Shah D, Shah D, Ndandji S, Kar S. Lazertinib: a novel EGFR-TKI therapy for non-small cell lung cancer. Expert Opin Drug Metab Toxicol 2025:1-12. [PMID: 40372786 DOI: 10.1080/17425255.2025.2507404] [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: 03/10/2025] [Revised: 04/17/2025] [Accepted: 05/13/2025] [Indexed: 05/17/2025]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) is the most prevalent form of lung cancer, accounting for 85% of cases worldwide. Despite advancements in treatment, many patients are diagnosed at advanced stages, and resistance to therapy, such as EGFR inhibitors, remains a significant challenge. Lazertinib, a third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) developed by Yuhan Corporation and Janssen Biotech, targets EGFR mutations, including T790M, which confer resistance to earlier-generation TKIs. AREAS COVERED This review explores lazertinib's development, mechanism of action, clinical efficacy, and safety profile. Preclinical studies demonstrated its superior selectivity for mutant EGFR and blood-brain barrier penetration compared to osimertinib. Clinical trials highlight its efficacy as monotherapy and in combination with amivantamab, showing improved progression-free survival and response duration in patients with advanced NSCLC. EXPERT OPINION Lazertinib represents a promising advance in the treatment of EGFR-mutated NSCLC, particularly for patients with brain metastases or resistance to previous EGFR TKIs. However, emerging resistance mutations, such as C797S, underscore the need for continued innovation, including combination therapies and fourth-generation TKIs. Future research must address these challenges to optimize treatment outcomes for NSCLC patients.
Collapse
Affiliation(s)
- Dhairavi Shah
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, Union, NJ, USA
| | - Dhaara Shah
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, Union, NJ, USA
| | - Suzy Ndandji
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, Union, NJ, USA
| | - Supratik Kar
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, Union, NJ, USA
| |
Collapse
|
4
|
Pozzetti L, Pinhammer MM, Asquith CRM. Medicinal chemistry applications of the Dimroth Rearrangement to the synthesis of biologically active compounds. Eur J Med Chem 2025; 289:117399. [PMID: 40024165 DOI: 10.1016/j.ejmech.2025.117399] [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: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
The Dimroth Rearrangement (DR) is an isomerization process involving the translocation of exo- and endocyclic nitrogen atoms in heterocyclic systems via a ring opening, rotation, and ring closure mechanism. Originally discovered over 120 years ago, the mechanistic occurrence of the DR on multiple heterocycles has been widely studied, and its application to the synthesis of biologically active compounds is well documented, albeit on some occasions not directly referenced. A surprisingly high number of drug discovery programs take advantage of the DR for the synthesis of heterocycle-containing compounds, including 4-aminopyrimidines and 4-anilinoquinazolines. Evidence of the flexibility and valuable potential of the DR can be found in the use of this reaction in the manufacture processes of several active pharmaceutical ingredients (APIs) on a commercial scale, allowing a reduction in the manufacturing costs and the environmental burden of the synthetic routes. The aim of this review is to outline the generality and broad applicability of the DR to the synthesis of biologically active compounds and highlight the opportunities to utilize this tool more widely within the medicinal chemistry toolbox.
Collapse
Affiliation(s)
- Luca Pozzetti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maja M Pinhammer
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Christopher R M Asquith
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
5
|
He P, Li H, Yang Z, Zhang R, Ye Q, Deng T, Li W, He S, Dong G, Yu Z, Li Y. Discovery and preclinical evaluations of drug candidate DA-0157 capable of overcoming EGFR drug-resistant mutation C797S and EGFR/ALK co-mutations. Eur J Med Chem 2025; 287:117323. [PMID: 39892095 DOI: 10.1016/j.ejmech.2025.117323] [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: 11/21/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Activating mutations in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) are significant oncogenic drivers in non-small cell lung cancer (NSCLC) patients. Despite several approved EGFR and ALK inhibitors, drug-resistant mutations pose a major challenge. Especially, there is currently no approved EGFR inhibitors targeting the C797S mutation, a refractory mutation resistant to the third-generation EGFR inhibitors. Furthermore, an increasing number of patients with EGFR/ALK co-mutations have been identified in clinical practice, yet there are no effective therapeutic options available for them. In this study, we report the discovery and preclinical evaluations of a new small-molecule drug candidate, DA-0157, which is capable of overcoming EGFR drug-resistant mutation C797S and EGFR/ALK co-mutations. DA-0157 demonstrated excellent in vitro efficacy, significantly inhibiting various EGFRC797S mutants resistant to the third-generation EGFR inhibitors, ALK rearrangements, and EGFR/ALK co-mutations. In vivo studies revealed that DA-0157 substantially inhibited tumor growth in the LD1-0025-200717 EGFRDel19/T790M/C797S PDX model (40 mg/kg/d, TGI: 98.3 %), Ba/F3-EML-4-ALK-L1196 M CDX model (40 mg/kg/d, TGI: 125.2 %), and NCI-H1975 EGFRDel19/T790M/C797S & NCI-H3122 (EML4-ALK) dual-side implantation CDX model (40 mg/kg/d, TGI: 89.5 % & 113.9 %). DA-0157 demonstrates favorable pharmacokinetic properties and safety. Currently, DA-0157 (DAJH-1050766) is undergoing Phase I/II clinical trials.
Collapse
Affiliation(s)
- Peng He
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Haiyan Li
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Zhenyu Yang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Qijun Ye
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Ta Deng
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Wenwen Li
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Shucheng He
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Guangxin Dong
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China.
| | - Zhou Yu
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China.
| | - Yi Li
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China.
| |
Collapse
|
6
|
Hou H, Liu X, Liu W, Zhang P, Zhou B. Warhead-bearing natural compounds for multi-pathway irreversible inhibition to overcome drug resistance in colorectal cancer. Med Oncol 2025; 42:148. [PMID: 40172739 DOI: 10.1007/s12032-025-02699-0] [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: 11/28/2024] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
Colorectal cancer (CRC) remains the second leading cause of cancer-related deaths globally, with approximately 930000 fatalities recorded in 2020. Resistance to conventional therapies continues to be a major obstacle in colorectal cancer treatment, highlighting the need for novel therapeutic strategies to enhance efficacy. This study aims to bridge this gap by exploring a multi-target inhibition approach using naturally derived electrophilic compounds, offering a potential solution to overcome drug resistance. Key CRC-covalent targets-EGFR, SRC, AKT1, HER2, and ERK2-were identified through network pharmacology and protein-protein interaction analysis. A panel of natural compounds, including ophiobolin A, deoxyelephantopin, eupalmerin acetate, curcumin, andrographolide, and syringolin A, was assessed for their inhibitory potential, benchmarking their activity against reference chemotherapeutics. Covalent docking and covalent molecular dynamics (CMD) were performed for 30 ligand-protein complexes to evaluate the binding affinities of the studied compounds. Against EGFR, curcumin displayed a competitive docking score of - 9.458 kcal/mol and ΔGbind of - 23.00 kcal/mol, closely matching the performance of afatinib (- 10.134 kcal/mol and - 24.28 kcal/mol, respectively). Syringolin A and andrographolide also exhibited strong binding affinities for EGFR. Against SRC, curcumin and andrographolide demonstrated excellent binding potential, achieving docking scores of - 8.360 and - 6.585 kcal/mol and ΔGbind values of - 38.91 and - 34.00 kcal/mol, respectively. In the case of AKT1, andrographolide displayed a competitive performance (- 8.044 kcal/mol, ΔGbind: - 32.00 kcal/mol), followed by curcumin and syringolin A. Andrographolide achieved the strongest binding affinity among the natural compounds against HER2 (- 7.006 kcal/mol, ΔGbind: - 21.01 kcal/mol) and ERK2 (- 7.640 kcal/mol, ΔGbind: - 33.00 kcal/mol), outperforming curcumin (- 7.468 kcal/mol, ΔGbind: - 31.23 kcal/mol) and deoxyelephantopin (- 6.517 kcal/mol, ΔGbind: - 29.01 kcal/mol). These results underscore the strong binding affinities of natural compounds to CRC targets and suggest that these compounds, either as standalone agents or in combination therapies, could complement existing chemotherapeutics by overcoming treatment resistance, thereby improving therapeutic outcomes in CRC patients.
Collapse
Affiliation(s)
- Huaping Hou
- Department of General Surgery, The First Hospital of Yulin, No. 93, Yuxi Avenue, Yuyang District, Yulin City, 719000, Shaanxi, China
| | - Xinqi Liu
- Department of General Surgery, The First Hospital of Yulin, No. 93, Yuxi Avenue, Yuyang District, Yulin City, 719000, Shaanxi, China
| | - Wentao Liu
- Surgery Department, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xi'an, 712000, China
| | - Pengfei Zhang
- Department of General Surgery, The First Hospital of Yulin, No. 93, Yuxi Avenue, Yuyang District, Yulin City, 719000, Shaanxi, China
| | - Bin Zhou
- Surgery Department, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xi'an, 712000, China.
| |
Collapse
|
7
|
Sun Z, Huo D, Guo J, Yan A. Modeling and Interpretability Study of the Structure-Activity Relationship for Multigeneration EGFR Inhibitors. ACS OMEGA 2025; 10:11176-11187. [PMID: 40160792 PMCID: PMC11947818 DOI: 10.1021/acsomega.4c10464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
The fourth-generation EGFR inhibitors targeting L858R/T790M/C797S mutations are in clinical trials mostly, and it is necessary to develop new inhibitors. In this study, an internal data set containing 2302 multitarget EGFR inhibitors targeting the wild type (83%) and the L858R (92%), L858R/T790M (96%), and L858R/T790M/C797S (60%) mutations was collected. We established a structure-activity relationship model for predicting the bioactivities of multigeneration EGFR inhibitors by a multitask deep neural network (MT-DNN). We also constructed four single-task models on 1384 L858R/T790M/C797S (60%) mutation inhibitors by support vector machine (SVM), random forest (RF), XGBoost (XGB), and single-target neural network (ST-DNN), respectively. The MT-DNN model significantly outperformed single-task models on the external data set of 304 fourth-generation EGFR inhibitors. Furthermore, the combined application of MT-DNN and SHAP/delta-SHAP value interpretability analysis offers rigorous structural information from a global perspective. With SHAP/delta-SHAP methods, the MT-DNN model can mine the core scaffold and important fragments of multigeneration EGFR inhibitors and provide valuable information from a structure-activity relationship perspective to address the resistant mutation problem.
Collapse
Affiliation(s)
- Zhiqi Sun
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| | - Donghui Huo
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| | - Jiangyu Guo
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| | - Aixia Yan
- State Key Laboratory of Chemical
Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East
Road, Beijing 100029, China
| |
Collapse
|
8
|
Lee CY, Lee SW, Hsu YC. Drug Resistance in Late-Stage Epidermal Growth Factor Receptor (EGFR)-Mutant Non-Small Cell Lung Cancer Patients After First-Line Treatment with Tyrosine Kinase Inhibitors. Int J Mol Sci 2025; 26:2042. [PMID: 40076686 PMCID: PMC11900297 DOI: 10.3390/ijms26052042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The development of tyrosine kinase inhibitors (TKIs) for late-stage epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) represented a drastic change in the treatment of late-stage lung cancer. Drug resistance develops after a certain period of first-line TKI treatment, which has led to decades of changing treatment guidelines for EGFR-mutant NSCLC. This study discussed the potential mechanisms of drug resistance against first-line TKI treatment and potential successive treatment strategies. Next-generation sequencing (NGS) may play a role in the evaluation of drug resistance in first-line TKI treatment. Emerging combination regimens and ongoing trials were discussed. Potential future strategies for treatment and for the management of drug resistance were proposed in this study.
Collapse
Affiliation(s)
- Ching-Yi Lee
- Department of Internal Medicine, Tao Yuan General Hospital, Taoyuan 33004, Taiwan;
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
| | - Shih-Wei Lee
- Department of Internal Medicine, Tao Yuan General Hospital, Taoyuan 33004, Taiwan;
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan
- Center for Astronautical Physics and Engineering, National Central University, Taoyuan 320317, Taiwan
- Department of Medical Research, Cathay General Hospital, Taipei 106438, Taiwan
| |
Collapse
|
9
|
Goodwin CR, De la Garza Ramos R, Bettegowda C, Barzilai O, Shreyaskumar P, Fehlings MG, Laufer I, Sahgal A, Rhines LD, Reynolds JJ, Lazary A, Gasbarrini A, Dea N, Verlaan JJ, Sullivan PZ, Gokaslan ZL, Fisher CG, Boriani S, Shin JH, Hornicek FJ, Weber MH, Goodwin ML, Charest-Morin R. Overview of Molecular Prognostication for Common Solid Tumor Histologies - What the Surgeon Should Know. Global Spine J 2025; 15:6S-15S. [PMID: 39801124 PMCID: PMC11726510 DOI: 10.1177/21925682241250327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
STUDY DESIGN Narrative Literature review. OBJECTIVE To provide a general overview of important molecular markers and targeted therapies for the most common neoplasms (lung, breast, prostate and melanoma) that metastasize to the spine and offer guidance on how to best incorporate them in the clinical setting. METHODS A narrative review of the literature was performed using PubMed, Google Scholar, Medline databases, as well as the histology-specific National Comprehensive Cancer Network guidelines to identify relevant articles limited to the English language. Relevant articles were reviewed for commonly described molecular mutations or targeted therapeutics, as well as associated clinical outcomes, and surgery-related risks. RESULTS Molecular markers and targeted therapies have dramatically improved the survival of cancer patients. The increasing importance of prognostic molecular markers and targeted therapies provides rationale for their incorporation into clinical decision-making for patients diagnosed with metastatic spine disease. In this review, we discuss the molecular markers/mutations and targeted therapies associated with the most common malignancies that metastasize to the spine and provide a framework that the surgeon can utilize when evaluating patients for potential intervention. Finally, we provide case examples that highlight the importance of molecular prognostication and therapies in surgical decision-making. CONCLUSION An integrated understanding of the implications of surgery, radiation, molecular markers and targeted therapies that guide prognostication and treatment is warranted in order to achieve the most favorable outcomes for patients with metastatic spine disease.
Collapse
Affiliation(s)
- C Rory Goodwin
- Department of Neurosurgery, Spine Division, Duke University, Durham, NC, USA
| | - Rafael De la Garza Ramos
- Department of Neurological Surgery, Montefiore Medical Center Albert Einstein College of Medicine, New York, NY, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ori Barzilai
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Patel Shreyaskumar
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Michael G Fehlings
- Department of Surgery, Division of Neurosurgery and Spine Program, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Ilya Laufer
- Department of Neurological Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Laurence D Rhines
- Department of Neurosurgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | | | - Aron Lazary
- National Center for Spinal Disorders, Budapest, Hungary
| | - Alessandro Gasbarrini
- Spine Surgery, University of Bologna, Contract Professor of Orthopedics at PostGraduate School, Bologna, Italy
| | - Nicolas Dea
- Combined Neurosurgical and Orthopedic Spine Program, Department of Orthopedics Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Jorrit-Jan Verlaan
- Department of Orthopaedic Surgery, University Medical Center Utrecht - Utrecht University, Utrecht, The Netherlands
| | - Patricia Zadnik Sullivan
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ziya L Gokaslan
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Charles G Fisher
- Combined Neurosurgical and Orthopedic Spine Program, Department of Orthopedics Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Stefano Boriani
- Spine Surgery, University of Bologna, Contract Professor of Orthopedics at PostGraduate School, Bologna, Italy
| | - John H Shin
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francis J Hornicek
- Department of Orthopaedic Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael H Weber
- Spine Surgery Program, Department of Surgery, Montreal General Hospital, McGill University Health Center, Montreal, QC, Canada
| | - Matthew L Goodwin
- Department of Orthopaedic Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Raphaële Charest-Morin
- Combined Neurosurgical and Orthopedic Spine Program, Department of Orthopedics Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Upadhyay R, Tandel P, Patel AB. Halogen-based quinazolin-4(3H)-one derivatives as MCF-7 breast cancer inhibitors: Current developments and structure-activity relationship. Arch Pharm (Weinheim) 2025; 358:e2400740. [PMID: 39535302 DOI: 10.1002/ardp.202400740] [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: 09/12/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Currently, cancer is a serious health challenge with predominance beyond restrictions. Breast cancer remains one of the major contributors to cancer-related morbidity and mortality in women. Chemotherapy continues to be crucial in the treatment of all variants of cancer. Several antitumor drugs are presently in different phases of clinical trials, whereas many more have been approved for clinical use. However, these drugs have the potential to cause adverse effects, and certain individuals may become resistant to them, which would eventually reduce the drug's efficacy. Therefore, it is essential to discover, develop, and improve newer anticancer drug molecules that could potentially inhibit proliferative pathways. In recent years, quinazolinone derivatives, more specifically halogen-substituted 4(3H)-quinazolinone, have drawn attention as a promising new class of chemotherapeutic agents. In addition, these molecules showed significant inhibition in micromolar ranges when tested in vitro against the MCF-7 cell line. Therefore, this study aims to emphasize the intriguing versatility of halogen atoms, providing an in-depth summary and highlighting recent developments in the anticancer properties of halogenated 4(3H)-quinazolinones. It also features a detailed discussion of the structure-activity relationship (SAR) of various functional groups and their interaction with amino acid residues utilizing molecular docking studies. The intent is to foster novel discoveries that can inspire innovative investigations in this domain. Hence, this study simplifies the drug design and development strategies by prolonging the array of pharmacologically active candidates.
Collapse
Affiliation(s)
- Rachana Upadhyay
- Department of Chemistry, Government College, Daman (Affiliated to Veer Narmad South Gujarat University, Surat), Daman, India
| | - Pooja Tandel
- Department of Chemistry, Government College, Daman (Affiliated to Veer Narmad South Gujarat University, Surat), Daman, India
| | - Amit B Patel
- Department of Chemistry, Government College, Daman (Affiliated to Veer Narmad South Gujarat University, Surat), Daman, India
| |
Collapse
|
11
|
Dou D, Zhang X, Wang J, Wumaier G, Qiao Y, Xie L, Jiang W, Sha W, Li W, Mei W, Zhang C, He H, Wang C, Wu L, Diao Y, Zhu L, Zhao Z, Chen Z, Xu Y, Li S, Li H. Design, synthesis, and biological evaluation of diphenyl ether substituted quinazolin-4-amine derivatives as potent EGFR L858R/T790M/C797S inhibitors. Eur J Med Chem 2024; 279:116858. [PMID: 39278125 DOI: 10.1016/j.ejmech.2024.116858] [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: 07/17/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Epidermal growth factor receptor (EGFR) is a validated target for non-small-cell lung cancer (NSCLC). However, the treatment for EGFR-C797S mutation induced by third-generation EGFR inhibitors remains a concern. Therefore, the development of the fourth-generation EGFR inhibitors to overcome the EGFR-C797S mutation has great potential for clinical treatment. In this article, we designed and synthesized a series of diphenyl ether substituted quinazolin-4-amine derivatives that simultaneously occupy the ATP binding pocket and the allosteric site of EGFR. Among the newly synthesized compounds, 9d displayed excellent kinase activity against EGFRL858R/T790M/C797S with an IC50 value of 0.005 μM, and exhibited anti-proliferation activity in BaF3-EGFRL858R/T790M/C797S cells with the IC50 value of 0.865 μM. Furthermore, 9d could suppress phosphorylation of EGFR and induce cell apoptosis and cycle arrest at G2 phase in a dose-dependent manner in BaF3-EGFRL858R/T790M/C797S cells. More importantly, 9d displayed significant antitumor effects in BaF3-EGFRL858R/T790M/C797S xenograft mouse model (30 mg/kg, TGI = 71.14 %). All the results indicated compound 9d might be a novel fourth-generation EGFR inhibitor for further development in overcoming the EGFR-C797S resistance mutation.
Collapse
Affiliation(s)
- Dou Dou
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, College of Life Sciences, Hebei Agricultural University, Baoding, 071001, China
| | - Xingsen Zhang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Jie Wang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Gulinuer Wumaier
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yunjin Qiao
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lijuan Xie
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenzhe Jiang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenjie Sha
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenjie Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenyi Mei
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Chen Zhang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Huan He
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Caolin Wang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lingkang Wu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yanyan Diao
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lili Zhu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhenjiang Zhao
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhuo Chen
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Yufang Xu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Shengqing Li
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
12
|
Martins DM, Fernandes PO, Vieira LA, Maltarollo VG, Moraes AH. Structure-Guided Drug Design Targeting Abl Kinase: How Structure and Regulation Can Assist in Designing New Drugs. Chembiochem 2024; 25:e202400296. [PMID: 39008807 DOI: 10.1002/cbic.202400296] [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: 03/31/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
The human protein Abelson kinase (Abl), a tyrosine kinase, plays a pivotal role in developing chronic myeloid leukemia (CML). Abl's involvement in various signaling pathways underscores its significance in regulating fundamental biological processes, including DNA damage responses, actin polymerization, and chromatin structural changes. The discovery of the Bcr-Abl oncoprotein, resulting from a chromosomal translocation in CML patients, revolutionized the understanding and treatment of the disease. The introduction of targeted therapies, starting with interferon-alpha and culminating in the development of tyrosine kinase inhibitors (TKIs) like imatinib, significantly improved patient outcomes. However, challenges such as drug resistance and side effects persist, indicating the necessity of research into novel therapeutic strategies. This review describes advancements in Abl kinase inhibitor development, emphasizing rational compound design from structural and regulatory information. Strategies, including bivalent inhibitors, PROTACs, and compounds targeting regulatory domains, promise to overcome resistance and minimize side effects. Additionally, leveraging the intricate structure and interactions of Bcr-Abl may provide insights into developing inhibitors for other kinases. Overall, this review highlights the importance of continued research into Abl kinase inhibition and its broader implications for therapeutic interventions targeting kinase-driven diseases. It provides valuable insights and strategies that may guide the development of next-generation therapies.
Collapse
MESH Headings
- Humans
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Drug Design
- Proto-Oncogene Proteins c-abl/metabolism
- Proto-Oncogene Proteins c-abl/antagonists & inhibitors
- Proto-Oncogene Proteins c-abl/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Molecular Structure
Collapse
Affiliation(s)
- Diego M Martins
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Philipe O Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Lucas A Vieira
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Vinícius G Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Adolfo H Moraes
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| |
Collapse
|
13
|
Zhao H, Beyett TS, Jiang J, Rana JK, Schaeffner IK, Santana J, Jänne PA, Eck MJ. Biochemical analysis of EGFR exon20 insertion variants insASV and insSVD and their inhibitor sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2417144121. [PMID: 39471218 PMCID: PMC11551396 DOI: 10.1073/pnas.2417144121] [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: 08/24/2024] [Accepted: 09/30/2024] [Indexed: 11/01/2024] Open
Abstract
Somatic mutations in the epidermal growth factor receptor (EGFR) are a major cause of non-small cell lung cancer. Among these structurally diverse alterations, exon 20 insertions represent a unique subset that rarely respond to EGFR tyrosine kinase inhibitors (TKIs). Therefore, there is a significant need to develop inhibitors that are active against this class of activating mutations. Here, we conducted biochemical analysis of the two most frequent exon 20 insertion variants, V769_D770insASV (insASV) and D770_N771insSVD (insSVD) to better understand their drug sensitivity and resistance. From kinetic studies, we found that EGFR insASV and insSVD are similarly active, but have lower Km, ATP values compared to the L858R variant, which contributes to their lack of sensitivity to 1st-3rd generation EGFR TKIs. Biochemical, structural, and cellular studies of a diverse panel of EGFR inhibitors revealed that the more recently developed compounds BAY-568, TAS6417, and TAK-788 inhibit EGFR insASV and insSVD in a mutant-selective manner, with BAY-568 being the most potent and selective versus wild-type (WT) EGFR. Cocrystal structures with WT EGFR reveal the binding modes of each of these inhibitors and of poziotinib, a potent but not mutantselective inhibitor, and together they define interactions shared by the mutant-selective agents. Collectively, our results show that these exon20 insertion variants are not inherently inhibitor resistant, rather they differ in their drug sensitivity from WT EGFR. However, they are similar to each other, indicating that a single inhibitor should be effective for several of the diverse exon 20 insertion variants.
Collapse
Affiliation(s)
- Hanchen Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
| | - Tyler S. Beyett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
| | - Jie Jiang
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
| | - Jaimin K. Rana
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Ilse K. Schaeffner
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Jhasmer Santana
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Pasi A. Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
| | - Michael J. Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
14
|
Abdel-Rahman AAH, El-Bayaa MN, Sobhy A, El-Ganzoury EM, Nossier ES, Awad HM, El-Sayed WA. Novel quinazolin-4-one based derivatives bearing 1,2,3-triazole and glycoside moieties as potential cytotoxic agents through dual EGFR and VEGFR-2 inhibitory activity. Sci Rep 2024; 14:24980. [PMID: 39443462 PMCID: PMC11500008 DOI: 10.1038/s41598-024-73171-8] [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: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
The toxicity that was caused by the developed medications for anticancer treatment is, unfortunately, an earnest problem stemming from the various involved targets, and accordingly, intense research for overcoming such a phenomenon remains indispensable. In the current inquiry, an innovative category of substituted quinazoline-based glycosides incorporating a core of 1,2,3-triazole and attached to distinct acetylated likewise deprotected sugar segments are created and produced synthetically. The resulted 1,2,3-triazolyl-glycosides products were investigated for their ability to cause cytotoxicity to several human cancer cell lines. The quinazoline based glycosyl-1,2,3-triazoles 10-13 with free hydroxy sugar moiety revealed excellent potency against (IC50 range = 5.70-8.10 µM, IC50 Doxorubicin = 5.6 ± 0.30 µM, IC50 Erlotinib = 4.3 ± 0.1 µM). against MCF-7 cancer cell line. In addition, the derived glycosides incorporating quinazolinone and triazole core 6-13 with acetylated and deprotected sugar parts showed excellent and superior potency against HCT-116 (IC50 range = 2.90-6.40 µM). The potent products were revealed as safe cytotoxic agents as indicated by their studied safety profiles. Additional research of promising candidates inhibitory analysis performed against EGFR and VEGFR-2. The hydroxylated glycosides incorporating triazole and quinazoline system 11 and 13 with N-methyl substitution of quinazolinone, gave excellent potency against EGFR (IC50 = 0.35 ± 0.11 and 0.31 ± 0.06 µM, correspondingly) since glycoside 13 revealed comparable IC50 (3.20 ± 0.15 µM) to sorafenib against VEGFR-2. For more understanding of its action mode, it was analyzed how the 1,2,3-triazolyl glycoside 13 made an effect on the apoptosis induction and the arrest of the cell cycle. It was revealed that it had the ability to stop HCT-116 cells in their cell cycle's G1 stage. Moreover, the influence of quinazolinone-1,2,3-triazole-glycoside 13 upon p53, Bax, and Bcl-2 levels in HCT-116 units was also studied for future approaches toward its behavior. Additionally, the latter derivative may trigger apoptosis, as indicated by a significant increase in apoptotic cells. Furthermore, molecular docking was simulated to make an obvious validation and comprehension acquirement of the binding's characteristics also attractions among the most forceful compounds side by side with their aimed enzymes.
Collapse
Affiliation(s)
- Adel A-H Abdel-Rahman
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom, Egypt.
| | - Mohamed N El-Bayaa
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Asmaa Sobhy
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom, Egypt
| | - Eman M El-Ganzoury
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom, Egypt
| | - Eman S Nossier
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt
- The National Committee of Drugs, Academy of Scientific Research and Technology, Cairo, 11516, Egypt
| | - Hanem M Awad
- Tanning Materials and Leather Technology Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Wael A El-Sayed
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| |
Collapse
|
15
|
Liang L, Zhang Z, You Q, Guo X. Recent advances in the design of small molecular drugs with acrylamides covalent warheads. Bioorg Med Chem 2024; 112:117902. [PMID: 39236467 DOI: 10.1016/j.bmc.2024.117902] [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: 06/11/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
In the development of covalent inhibitors, acrylamides warhead is one of the most popular classes of covalent warheads. In recent years, researchers have made different structural modifications to acrylamides warheads, resulting in the creation of fluorinated acrylamide warheads and cyano acrylamide warheads. These new warheads exhibit superior selectivity, intracellular accumulation, and pharmacokinetic properties. Additionally, although ketoamide warheads have been applied in the design of covalent inhibitors for viral proteins, it has not received sufficient attention. Combined with the studies in kinase inhibitors and antiviral drugs, this review presents the structural features and the progression of acrylamides warheads, offering a perspective on future research and development in this field.
Collapse
Affiliation(s)
- Luxia Liang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ze Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
16
|
Ni H, Wang Z, Tang Y, Lu J, Zhu Z, Qiu Y, Chen Z, Wang Z. Tyrosine kinase inhibitors in the treatment of leptomeningeal carcinomatosis. Cell Biol Int 2024; 48:1450-1462. [PMID: 39136350 DOI: 10.1002/cbin.12230] [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: 12/28/2023] [Revised: 06/28/2024] [Accepted: 07/31/2024] [Indexed: 10/19/2024]
Abstract
Leptomeningeal carcinomatosis (LMC) is a devastating complication of advanced cancers, such as lung cancer and breast cancer, which is usually indicative of a poor prognosis. The current treatments for LMC include palliative care, with others aiming to prolong survival and relieve neurological symptoms. Traditional treatments for LMC include radiotherapy, systemic chemotherapy, and intrathecal injection. Furthermore, the application of molecularly targeted agents, such as antiepidermal growth factor receptor (anti-EGFR), antihuman epidermal growth factor receptor 2 (anti-HER2), and anti-PD-1 monoclonal antibody, have prolonged the survival of LMC patients. Targeted therapy with tyrosine kinase inhibitors has also been proven to be an effective treatment. Tyrosine kinases can be overactive or expressed at high levels in some cancer cells; therefore, the use of tyrosine kinase inhibitors may prevent the activation of tumor-related pathways, preventing cancer cell growth. The EGFR family are cell surface receptors directly related to tumor occurrence with tyrosine kinase activity; it is the most widely used target for tyrosine kinase inhibitors in the treatment of LMC. In this review, we introduced the clinical manifestation and diagnostic criteria of LMC, clarified the treatment mechanism of tyrosine kinase inhibitors for LMC with mutations in EGFR, HER2, or anaplastic lymphoma kinase, reviewed the current application of various generation tyrosine kinase inhibitors in patients with LMC, and discussed new clinical trials and the future directions of tyrosine kinase inhibitor therapy.
Collapse
Affiliation(s)
- Hanyu Ni
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Zilan Wang
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanbing Tang
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiaye Lu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zixiang Zhu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Youjia Qiu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhouqing Chen
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
17
|
Dickerson H, Diab A, Al Musaimi O. Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Cancer: Current Use and Future Prospects. Int J Mol Sci 2024; 25:10008. [PMID: 39337496 PMCID: PMC11432255 DOI: 10.3390/ijms251810008] [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: 08/30/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have emerged as a leading targeted cancer therapy, reducing the side effects often seen with non-targeted treatments, especially the damage to healthy cells. To tackle resistance, typically caused by epidermal growth factor receptor (EGFR) mutations, four generations of TKIs have been developed. Each generation has shown improved effectiveness and fewer side effects, resulting in better patient outcomes. For example, patients on gefitinib, a first-generation TKI, experienced a progression-free survival (PFS) of 10 months compared to 5 months with conventional chemotherapy. Second-generation TKI afatinib outperformed erlotinib and extended PFS to 11.1 months compared to 6.9 months with cisplatin. Third-generation TKIs further increased survival to 38.6 months, compared to 31.8 months with first-generation TKIs. This progress demonstrates the ability of newer TKIs to overcome resistance, particularly the T790M mutation, while reducing adverse effects. Ongoing research focuses on overcoming resistance from newer mutations like C797S to further improve patient survival. These developments highlight the significant progress in TKI therapy and the continued effort to refine cancer treatment. Recent research in South Korea shows that third-generation TKIs are ineffective against non-small cell lung cancer (NSCLC) with the C797S mutation. Several trials have started showing promising in vitro and in vivo results, but more trials are needed before clinical approval. This review underscores notable advancements in the field of EGFR TKIs, offering a comprehensive analysis of their mechanisms of action and the progression of various TKI generations in response to resistance.
Collapse
Affiliation(s)
- Henry Dickerson
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Ahmad Diab
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Othman Al Musaimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
18
|
El-Bayaa MN, Kotb ER, Messaoudi S, Awad HM, Saleh MG, Soliman HA. Synthesis, anticancer activity, docking and computational studies of new pyridyl-glycosyl hybrids and acyclic analogs. Future Med Chem 2024; 16:2119-2133. [PMID: 39263964 PMCID: PMC11559364 DOI: 10.1080/17568919.2024.2389768] [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: 05/30/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024] Open
Abstract
New pyridine-O-glycosides and their acyclic nucleoside analogues were prepared by heterocyclization and glycosylation. The anticancer activity against HCT-116, HepG2 and MCF-7 human cancer cells and BJ-1 cell revealed that the galacto- and xylopyranosyl glycosides possessing 4-bromophenyl have superior cytotoxic activities against HepG2 cell while glycosides 7-9 resulted in superior cytotoxic activities regarding MCF-7 breast cell. In case of HCT-116 colorectal carcinoma cells, two products and the derived glycosides and acyclic analogues showed potent activities. The most potent compounds were investigated for their possible binding affinities to the active site of CDK2 enzyme via in silico molecular docking simulation in addition to computational studies. The results support the antiproliferative effect and elucidate the interactions of 3a and 8 with catalytic sites.
Collapse
Affiliation(s)
- Mohamed N El-Bayaa
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Eman R Kotb
- Photochemistry Department, National Research Centre, Dokki, P.O. Box 12622, Cairo, Egypt
| | - Sabri Messaoudi
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Hanem M Awad
- Tanning Materials & Leather Technology Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Mahmoud G Saleh
- Department of Chemistry, College of Science, Northern Border University, Arar, Saudi Arabia
| | - Hanan A Soliman
- Photochemistry Department, National Research Centre, Dokki, P.O. Box 12622, Cairo, Egypt
| |
Collapse
|
19
|
Rocha KML, Nascimento ÉCM, de Jesus RCC, Martins JBL. In Silico Molecular Modeling of Four New Afatinib Derived Molecules Targeting the Inhibition of the Mutated Form of BCR-ABL T315I. Molecules 2024; 29:4254. [PMID: 39275102 PMCID: PMC11397288 DOI: 10.3390/molecules29174254] [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: 06/12/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/16/2024] Open
Abstract
Four afatinib derivatives were designed and modeled. These derivatives were compared to the known tyrosine-kinase inhibitors in treating Chronic Myeloid Leukemia, i.e., imatinib and ponatinib. The molecules were evaluated through computational methods, including docking studies, the non-covalent interaction index, Electron Localization and Fukui Functions, in silico ADMET analysis, QTAIM, and Heat Map analysis. The AFA(IV) candidate significantly increases the score value compared to afatinib. Furthermore, AFA(IV) was shown to be relatively similar to the ponatinib profile when evaluating a range of molecular descriptors. The addition of a methylpiperazine ring seems to be well distributed in the structure of afatinib when targeting the BCR-ABL enzyme, providing an important hydrogen bond interaction with the Asp381 residue of the DFG-switch of BCR-ABL active site residue and the AFA(IV) new chemical entities. Finally, in silico toxicity predictions show a favorable index, with some molecules presenting the loss of the irritant properties associated with afatinib in theoretical predictions.
Collapse
MESH Headings
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/chemistry
- Afatinib/chemistry
- Afatinib/pharmacology
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Molecular Docking Simulation
- Humans
- Models, Molecular
- Computer Simulation
- Mutation
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Hydrogen Bonding
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Imidazoles/chemistry
- Imidazoles/pharmacology
- Pyridazines
Collapse
Affiliation(s)
- Kelvyn M. L. Rocha
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (K.M.L.R.); (É.C.M.N.); (R.C.C.d.J.)
| | - Érica C. M. Nascimento
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (K.M.L.R.); (É.C.M.N.); (R.C.C.d.J.)
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Rafael C. C. de Jesus
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (K.M.L.R.); (É.C.M.N.); (R.C.C.d.J.)
| | - João B. L. Martins
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (K.M.L.R.); (É.C.M.N.); (R.C.C.d.J.)
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasília, Brasília 70910-900, DF, Brazil
| |
Collapse
|
20
|
Lin Y, Huang Y, Yang B, Zhang Y, Ji N, Li J, Zhou Y, Shen YQ, Chen Q. Precision therapy targeting CAMK2 to overcome resistance to EGFR inhibitors in FAT1-mutated oral squamous cell carcinoma. Chin Med J (Engl) 2024:00029330-990000000-01204. [PMID: 39227322 DOI: 10.1097/cm9.0000000000003217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a prevalent type of cancer with a high mortality rate in its late stages. One of the major challenges in OSCC treatment is the resistance to epidermal growth factor receptor (EGFR) inhibitors. Therefore, it is imperative to elucidate the mechanism underlying drug resistance and develop appropriate precision therapy strategies to enhance clinical efficacy. METHODS To evaluate the efficacy of the combination of the Ca2+/calmodulin-dependent protein kinase II (CAMK2) inhibitor KN93 and EGFR inhibitors, we performed in vitro and in vivo experiments using two FAT atypical cadherin 1 (FAT1)-deficient (SCC9 and SCC25) and two FAT1 wild-type (SCC47 and HN12) OSCC cell lines. We assessed the effects of EGFR inhibitors (afatinib or cetuximab), KN93, or their combination on the malignant phenotype of OSCC in vivo and in vitro. The alterations in protein expression levels of members of the EGFR signaling pathway and SRY-box transcription factor 2 (SOX2) were analyzed. Changes in the yes-associated protein 1 (YAP1) protein were characterized. Moreover, we analyzed mitochondrial dysfunction. Besides, the effects of combination therapy on mitochondrial dynamics were also evaluated. RESULTS OSCC with FAT1 mutations exhibited resistance to EGFR inhibitors treatment. The combination of KN93 and EGFR inhibitors significantly inhibited the proliferation, survival, and migration of FAT1-mutated OSCC cells and suppressed tumor growth in vivo. Mechanistically, combination therapy enhanced the therapeutic sensitivity of FAT1-mutated OSCC cells to EGFR inhibitors by modulating the EGFR pathway and downregulated tumor stemness-related proteins. Furthermore, combination therapy induced reactive oxygen species (ROS)-mediated mitochondrial dysfunction and disrupted mitochondrial dynamics, ultimately resulting in tumor suppression. CONCLUSION Combination therapy with EGFR inhibitors and KN93 could be a novel precision therapeutic strategy and a potential clinical solution for EGFR-resistant OSCC patients with FAT1 mutations.
Collapse
Affiliation(s)
- Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bindra S, Bose K, Thekkantavida AC, Grace Thomas Parambi D, Alsahli TG, Pant M, Pappachen LK, Kim H, Mathew B. FDA-approved drugs containing dimethylamine pharmacophore: a review of the last 50 years. RSC Adv 2024; 14:27657-27696. [PMID: 39224646 PMCID: PMC11367245 DOI: 10.1039/d4ra04730c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Dimethylamine (DMA) derivatives represent a promising class of compounds with significant potential in the field of medicinal chemistry. DMA derivatives exhibit a diverse range of pharmacological activities, including antimicrobial, antihistaminic, anticancer, and analgesic properties. Their unique chemical structure allows for the modulation of various biological targets, making them valuable candidates for the treatment of numerous diseases. Synthetic strategies for the preparation of DMA derivatives vary depending on the desired biological activity and target molecule. Common synthetic routes involve the modification of the DMA scaffold through functional group manipulation, scaffold hopping, or combinatorial chemistry approaches. Therapeutically, DMA derivatives have shown promise in the treatment of infectious diseases, especially bacterial infections. Additionally, by focusing on particular biochemical pathways involved in tumor growth and metastasis, DMA-based drugs have shown anticancer activity. In addition to their direct pharmacological effects, DMA derivatives can serve as valuable tools in drug delivery systems, prodrug design, and molecular imaging techniques, enhancing their utility in medicinal chemistry research. Overall, DMA derivatives represent a versatile class of compounds with immense potential in medicinal chemistry. Further research and development efforts are warranted to explore their full therapeutic capabilities and optimize their clinical utility in the treatment of various diseases. This article outlines the pharmacological properties, synthetic strategies, and therapeutic applications of DMA derivatives of FDA approved drugs, highlighting their importance in drug discovery and development.
Collapse
Affiliation(s)
- Sandeep Bindra
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham Amrita Health Science Campus Kochi 682041 India
| | - Kuntal Bose
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham Amrita Health Science Campus Kochi 682041 India
| | - Amrutha Chandran Thekkantavida
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham Amrita Health Science Campus Kochi 682041 India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka Aljouf 72341 Saudi Arabia
| | - Tariq G Alsahli
- Department of Pharmacology, College of Pharmacy, Jouf University Sakaka Aljouf 72341 Saudi Arabia
| | - Manu Pant
- School of Pharmacy, Graphic Era Hill University Dehradun 248002 India
- Graphic Era Hill University (Deemed to be University) Clement Town Dehradun 248002 India
| | - Leena K Pappachen
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham Amrita Health Science Campus Kochi 682041 India
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University Suncheon 57922 Republic of Korea
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham Amrita Health Science Campus Kochi 682041 India
| |
Collapse
|
22
|
Elshazly AM, Xu J, Melhem N, Abdulnaby A, Elzahed AA, Saleh T, Gewirtz DA. Is Autophagy Targeting a Valid Adjuvant Strategy in Conjunction with Tyrosine Kinase Inhibitors? Cancers (Basel) 2024; 16:2989. [PMID: 39272847 PMCID: PMC11394573 DOI: 10.3390/cancers16172989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) represent a relatively large class of small-molecule inhibitors that compete with ATP for the catalytic binding site of tyrosine kinase proteins. While TKIs have demonstrated effectiveness in the treatment of multiple malignancies, including chronic myelogenous leukemia, gastrointestinal tumors, non-small cell lung cancers, and HER2-overexpressing breast cancers, as is almost always the case with anti-neoplastic agents, the development of resistance often imposes a limit on drug efficacy. One common survival response utilized by tumor cells to ensure their survival in response to different stressors, including anti-neoplastic drugs, is that of autophagy. The autophagic machinery in response to TKIs in multiple tumor models has largely been shown to be cytoprotective in nature, although there are a number of cases where autophagy has demonstrated a cytotoxic function. In this review, we provide an overview of the literature examining the role that autophagy plays in response to TKIs in different preclinical tumor model systems in an effort to determine whether autophagy suppression or modulation could be an effective adjuvant strategy to increase efficiency and/or overcome resistance to TKIs.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Nebras Melhem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan;
| | - Alsayed Abdulnaby
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Aya A. Elzahed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan;
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
| |
Collapse
|
23
|
Kumar M, Patil KT, Maity P, Chatterjee J, Singh T, Joshi G, Singh S, Kumar R. Design, synthesis, and anticancer assessment of structural analogues of ( E)-1-((3,4,5-trimethoxybenzylidene)amino)-4-(3,4,5-trimethoxyphenyl)imidazo[1,2- a]quinoxaline-2-carbonitrile (6b), an imidazo[1,2- a]quinoxaline-based non-covalent EGFR inhibitor. RSC Med Chem 2024; 15:2322-2339. [PMID: 39026650 PMCID: PMC11253857 DOI: 10.1039/d4md00237g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/26/2024] [Indexed: 07/20/2024] Open
Abstract
In our quest to find improved anticancer therapeutics, we expedite the lead optimization of (E)-1-((3,4,5-trimethoxybenzylidene)amino)-4-(3,4,5-trimethoxyphenyl)imidazo[1,2-a]quinoxaline-2-carbonitrile (6b), an EGFR inhibitor previously discovered in our laboratory through an in-house screening program. The lead optimization was rationally initiated considering the catalytic site of EGFR. We synthesized twenty-nine new analogues of 6b and assessed their anticancer activities. SAR studies highlighted the role of important groups in controlling anticancer activities. Among all, 5a and 5l were found to exhibit improved EGFR inhibition with anticancer asset potential. In silico studies corroborated with in vitro EGFR inhibitory results. The deeper analysis of 5a and 5l revealed that these synthetics could alter the MMP (ΔΨ m) and significantly reduce the ROS levels in lung cancer cells. This is a vital prerequisite for better plausible EGFR inhibitors devoid of cardiotoxicity. qPCR analysis further revealed that the investigational compounds 5a and 5l were able to downregulate the expression of key oncogenes, viz., KRAS, MAP2K, and EGFR. The downregulation of these genes suggests that the investigational compounds could interact and inhibit key players in the signalling cascade along with the EGFR, which may lead to the inhibition of the growth and prognosis of cancer cells via a holistic approach.
Collapse
Affiliation(s)
- Manvendra Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Kiran T Patil
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Pritam Maity
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Joydeep Chatterjee
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda - 151401 Punjab India
| | - Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal (A Central) University Dist. Garhwal Srinagar 246174 Uttarakhand India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda - 151401 Punjab India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| |
Collapse
|
24
|
Abdel-Aziz AAM, El-Azab AS, Brogi S, Ayyad RR, Alkahtani HM, Abuelizz HA, Al-Suwaidan IA, Al-Obaid AM. Synthesis, enzyme inhibition assay, and molecular modeling study of novel pyrazolines linked to 4-methylsulfonylphenyl scaffold: antitumor activity and cell cycle analysis. RSC Adv 2024; 14:22132-22146. [PMID: 39005246 PMCID: PMC11240878 DOI: 10.1039/d4ra03902e] [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: 05/27/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Antitumor activity using 59 cancer cell lines and enzyme inhibitory activity of a newly synthesized pyrazoline-linked 4-methylsulfonylphenyl scaffold (compounds 18a-q) were measured and compared with those of standard drugs. Pyrazolines 18b, 18c, 18f, 18g, 18h, and 18n possessed significant antitumor activity, with a positive cytotoxic effect (PCE) of 22/59, 21/59, 21/59, 48/59, 51/59, and 20/59, respectively. The cancer cell lines HL60, MCF-7, and MDA-MB-231 were used to measure the IC50 values of derivatives 18c, 18g, and 18hvia the MTT assay method, and the results were compared with those of reference drugs. Derivatives 18g and 18h showed potent and broad-spectrum antitumor activities against HL60 (IC50 of 10.43, 8.99 μM, respectively), MCF-7 (IC50 of 11.7 and 12.4 μM, respectively), and MDA-MB-231 (IC50 of 4.07 and 7.18 μM, respectively). Compound 18c exhibited strong antitumor activity against HL60 and MDA-MB-231 cell lines with IC50 values of 8.43 and 12.54 μM, respectively, and moderate antitumor activity against MCF-7 cell lines with an IC50 value of 16.20 μM. Compounds 18c, 18g, and 18h remarkably inhibited VEGFR2 kinase (IC50 = 0.218, 0.168, and 0.135 μM, respectively) compared with the reference drug sorafenib (IC50 = 0.041 μM). Compounds 18g and 18h effectively inhibited HER2 kinase (IC50 = 0.496 and 0.253 μM, respectively) compared with erlotinib (IC50 = 0.085 μM). Compound 18h inhibited EGFR kinase (IC50 = 0.574 μM) with a potency comparable with that of the reference drug erlotinib (IC50 = 0.105 μM). Pyrazolines 18c, 18f, and 18h arrested the S/G2 phase of the cell cycle in HL-60 cells. In addition, derivatives 18c, 18f, and 18h revealed lower Bcl-2 protein expression anti-apoptotic levels and higher Bax, caspase-3, and caspase-9 expression levels. Molecular docking studies of derivative 18h into the binding sites of EGFR, HER2, and VEGFR2 kinases explored the interaction mode of these pyrazoline derivatives and their structural requirements for antitumor activity.
Collapse
Affiliation(s)
- Alaa A-M Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Adel S El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Simone Brogi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Rezk R Ayyad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-AzharUniversity Cairo Egypt
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Hatem A Abuelizz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Ibrahim A Al-Suwaidan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Abdulrahman M Al-Obaid
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| |
Collapse
|
25
|
Shi JT, Hou SJ, Cheng L, Zhang HJ, Mu HX, Wang QS, Wang ZY, Chen SW. Discovery of novel coumarin-based KRAS-G12C inhibitors from virtual screening and Rational structural optimization. Bioorg Chem 2024; 148:107467. [PMID: 38772290 DOI: 10.1016/j.bioorg.2024.107467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
KRAS-G12C inhibitors has been made significant progress in the treatment of KRAS-G12C mutant cancers, but their clinical application is limited due to the adaptive resistance, motivating development of novel structural inhibitors. Herein, series of coumarin derivatives as KRAS-G12C inhibitors were found through virtual screening and rational structural optimization. Especially, K45 exhibited strong antiproliferative potency on NCI-H23 and NCI-H358 cancer cells harboring KRAS-G12C with the IC50 values of 0.77 μM and 1.50 μM, which was 15 and 11 times as potent as positive drug ARS1620, respectively. Furthermore, K45 reduced the phosphorylation of KRAS downstream effectors ERK and AKT by reducing the active form of KRAS (KRAS GTP) in NCI-H23 cells. In addition, K45 induced cell apoptosis by increasing the expression of anti-apoptotic protein BAD and BAX in NCI-H23 cells. Docking studies displayed that the 3-naphthylmethoxy moiety of K45 extended into the cryptic pocket formed by the residues Gln99 and Val9, which enhanced the interaction with the KRAS-G12C protein. These results indicated that K45 was a potent KRAS-G12C inhibitor worthy of further study.
Collapse
Affiliation(s)
- Jian-Tao Shi
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Su-Juan Hou
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Lei Cheng
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Hao-Jie Zhang
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Hong-Xia Mu
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Qing-Shan Wang
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Zhao-Yang Wang
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China
| | - Shi-Wu Chen
- School of Pharmacy & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
26
|
Manhas R, Rathore A, Havelikar U, Mahajan S, Gandhi SG, Mahapa A. Uncovering the potentiality of quinazoline derivatives against Pseudomonas aeruginosa with antimicrobial synergy and SAR analysis. J Antibiot (Tokyo) 2024; 77:365-381. [PMID: 38514856 DOI: 10.1038/s41429-024-00717-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/10/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Antimicrobial resistance has emerged as a covert global health crisis, posing a significant threat to humanity. If left unaddressed, it is poised to become the foremost cause of mortality worldwide. Among the multitude of resistant bacterial pathogens, Pseudomonas aeruginosa, a Gram-negative, facultative bacterium, has been responsible for mild to deadly infections. It is now enlisted as a global critical priority pathogen by WHO. Urgent measures are required to combat this formidable pathogen, necessitating the development of novel anti-pseudomonal drugs. To confront this pressing issue, we conducted an extensive screening of 3561 compounds from the ChemDiv library, resulting in the discovery of potent anti-pseudomonal quinazoline derivatives. Among the identified compounds, IDD-8E has emerged as a lead molecule, exhibiting exceptional efficacy against P. aeruginosa while displaying no cytotoxicity. Moreover, IDD-8E demonstrated significant pseudomonal killing, disruption of pseudomonal biofilm and other anti-bacterial properties comparable to a well-known antibiotic rifampicin. Additionally, IDD-8E's synergy with different antibiotics further strengthens its potential as a powerful anti-pseudomonal agent. IDD-8E also exhibited significant antimicrobial efficacy against other ESKAPE pathogens. Moreover, we elucidated the Structure-Activity-Relationship (SAR) of IDD-8E targeting the essential WaaP protein in P. aeruginosa. Altogether, our findings emphasize the promise of IDD-8E as a clinical candidate for novel anti-pseudomonal drugs, offering hope in the battle against antibiotic resistance and its devastating impact on global health.
Collapse
Affiliation(s)
- Rakshit Manhas
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Arti Rathore
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ujwal Havelikar
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Shavi Mahajan
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Sumit G Gandhi
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Avisek Mahapa
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
27
|
Li J, Gong C, Zhou H, Liu J, Xia X, Ha W, Jiang Y, Liu Q, Xiong H. Kinase Inhibitors and Kinase-Targeted Cancer Therapies: Recent Advances and Future Perspectives. Int J Mol Sci 2024; 25:5489. [PMID: 38791529 PMCID: PMC11122109 DOI: 10.3390/ijms25105489] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Over 120 small-molecule kinase inhibitors (SMKIs) have been approved worldwide for treating various diseases, with nearly 70 FDA approvals specifically for cancer treatment, focusing on targets like the epidermal growth factor receptor (EGFR) family. Kinase-targeted strategies encompass monoclonal antibodies and their derivatives, such as nanobodies and peptides, along with innovative approaches like the use of kinase degraders and protein kinase interaction inhibitors, which have recently demonstrated clinical progress and potential in overcoming resistance. Nevertheless, kinase-targeted strategies encounter significant hurdles, including drug resistance, which greatly impacts the clinical benefits for cancer patients, as well as concerning toxicity when combined with immunotherapy, which restricts the full utilization of current treatment modalities. Despite these challenges, the development of kinase inhibitors remains highly promising. The extensively studied tyrosine kinase family has 70% of its targets in various stages of development, while 30% of the kinase family remains inadequately explored. Computational technologies play a vital role in accelerating the development of novel kinase inhibitors and repurposing existing drugs. Recent FDA-approved SMKIs underscore the importance of blood-brain barrier permeability for long-term patient benefits. This review provides a comprehensive summary of recent FDA-approved SMKIs based on their mechanisms of action and targets. We summarize the latest developments in potential new targets and explore emerging kinase inhibition strategies from a clinical perspective. Lastly, we outline current obstacles and future prospects in kinase inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.L.)
| |
Collapse
|
28
|
Duan L, Chu C, Huang X, Yao H, Wen J, Chen R, Wang C, Tu Y, Lv Q, Pan Q, Xu S. Rational design and synthesis of 2,4-dichloro-6-methyl pyrimidine derivatives as potential selective EGFR T790M/L858R inhibitors for the treatment of non-small cell lung cancer. Arch Pharm (Weinheim) 2024; 357:e2300736. [PMID: 38381049 DOI: 10.1002/ardp.202300736] [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: 12/14/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/22/2024]
Abstract
Many patients with non-small cell lung cancer (NSCLC) initially benefit from epidermal growth factor receptor (EGFR) targeted therapy. Unfortunately, varying degrees of resistance or side effects eventually develop. Overcoming and preventing the resistance and side effects of EGFR inhibitors has become a hot topic of research today. Based on the previous studies on AZD-9291, we designed and synthesized two series of 2,4-dichloro-6-methylpyrimidine derivatives, 19 compounds in total, as potential inhibitors of the EGFR kinase. The most promising compound, L-18, showed better inhibitory activity (81.9%) and selectivity against EGFRT790M/L858R kinase. In addition, L-18 showed strong antiproliferative activity against H1975 cells with an IC50 value of 0.65 ± 0.06 μM and no toxicity to normal cells (LO-2). L-18 was able to dose-dependently induce the apoptosis of H1975 cells and produced a cell-cycle-blocking effect, and it can also dose-dependently inhibit the migration and invasion of H1975 cells. L-18 also showed in vivo anticancer efficacy in H1975 cells xenograft mice. We also performed a series of in vivo and in vitro toxicological evaluations of compound L-18, which did not cause obvious injury in mice during administration. These results suggest that L-18 may be a promising drug candidate that warrants further investigation.
Collapse
Affiliation(s)
- Lei Duan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Cilong Chu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Xiaoling Huang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Huizhi Yao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Jie Wen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Rui Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Caolin Wang
- School of Pharmacy, East China University of Science & Technology, Shanghai, China
| | - Yuanbiao Tu
- Cancer Research Center, Jangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qiaoli Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, People's Republic of China
| | - Qingshan Pan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| |
Collapse
|
29
|
Liu J, Nie W, Nie H, Yao H, Ren Y, Cao L, Qiu J, Wang M, Li X, An B, Jia X. The new N 2, N 4-diphenylpyridine-2,4-diamine deuterated derivatives as EGFR inhibitors to overcome C797S-mediated resistance. Bioorg Chem 2024; 146:107313. [PMID: 38554675 DOI: 10.1016/j.bioorg.2024.107313] [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: 02/04/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
A series of new deuterated and non-deuterated N2, N4-diphenylpyridine - 2,4-diamine derivatives were synthesized and evaluated as EGFR C797S-mediated resistance inhibitors. Most of these compounds exhibited potent antiproliferative activity against Baf3-EGFR L858R/T790M/C797S and Baf3-EGFR Del19/T790M/C797S cancel cell lines, with IC50 values in the nanomolar concentration range. Among them, compound 14l represented the most active compound with IC50 values of 8-11 nM. Interestingly, metabolic stability assay with rat liver microsomes indicated that the half-life of the deuterated derivative 14o was significantly increased compared to that of 14l. In xenograft mice models, 14o inhibited tumor growth with excellent inhibitory rate of 75.1 % at the dosage of 40 mg/kg, comparing 73.2 % of the TGI with its non-deuterated compound 14l, at a dosage of 80 mg/kg. Mechanism studies revealed that 14o was a potent EGFR L858R/T790M/C797S and EGFR Del19/T790M/C797S kinase inhibitor, which could downregulate the protein phosphorylation of EGFR and m-TOR signaling pathways, arrest cell cycle at G2/M phase by affecting the expression of CDC25C, and promote cell apoptosis by regulating the expression of cleaved caspase-3. In summary, 14o could serve as a promising deuterated compound for the development of highly efficient anticancer agents.
Collapse
Affiliation(s)
- Jiadai Liu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Wenyan Nie
- School of Pharmacy, Binzhou Medical University, Yantai 264003, PR China
| | - Haoran Nie
- School of Pharmacy, Binzhou Medical University, Yantai 264003, PR China
| | - Han Yao
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou 510990, PR China
| | - Yuanyuan Ren
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou 510990, PR China
| | - Longcai Cao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jiaqi Qiu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, PR China
| | - Mengxuan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, PR China
| | - Xingshu Li
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou 510990, PR China
| | - Baijiao An
- School of Pharmacy, Binzhou Medical University, Yantai 264003, PR China.
| | - Xian Jia
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
30
|
Acharya B, Saha D, Armstrong D, Jabali B, Hanafi M, Herrera-Rueda A, Lakkaniga NR, Frett B. Kinase inhibitor macrocycles: a perspective on limiting conformational flexibility when targeting the kinome with small molecules. RSC Med Chem 2024; 15:399-415. [PMID: 38389874 PMCID: PMC10880908 DOI: 10.1039/d3md00457k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/10/2023] [Indexed: 02/24/2024] Open
Abstract
Methods utilized for drug discovery and development within the kinome have rapidly evolved since the approval of imatinib, the first small molecule kinase inhibitor. Macrocycles have received increasing interest as a technique to improve kinase inhibitor drug properties evident by the FDA approvals of lorlatinib, pacritinib, and repotrectinib. Compared to their acyclic counterparts, macrocycles can possess improved pharmacodynamic and pharmacokinetic properties. This review highlights clinical success stories when implementing macrocycles in kinase-based drug discovery and showcases that macrocyclization is a clinically validated drug discovery strategy when targeting the kinome.
Collapse
Affiliation(s)
- Baku Acharya
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
| | - Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
- Conrad Prebys Centre for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute San Diego CA USA
| | - Daniel Armstrong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
| | - Baha'a Jabali
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
| | - Maha Hanafi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University Cairo 11526 Egypt
| | - Alan Herrera-Rueda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
| | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR USA
| |
Collapse
|
31
|
Serafim RAM, Gehringer M, Borsari C. Targeted Covalent Inhibitors in Drug Discovery, Chemical Biology and Beyond. Pharmaceuticals (Basel) 2024; 17:206. [PMID: 38399421 PMCID: PMC10891537 DOI: 10.3390/ph17020206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Covalent inhibitors have experienced a revival in medicinal chemistry and chemical biology in recent decades [...].
Collapse
Affiliation(s)
- Ricardo A. M. Serafim
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Matthias Gehringer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided & Functionally Instructed Tumor Therapies’, University of Tübingen, 72076 Tübingen, Germany
| | - Chiara Borsari
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
32
|
Frumento D, Grossi G, Falesiedi M, Musumeci F, Carbone A, Schenone S. Small Molecule Tyrosine Kinase Inhibitors (TKIs) for Glioblastoma Treatment. Int J Mol Sci 2024; 25:1398. [PMID: 38338677 PMCID: PMC10855061 DOI: 10.3390/ijms25031398] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
In the last decade, many small molecules, usually characterized by heterocyclic scaffolds, have been designed and synthesized as tyrosine kinase inhibitors (TKIs). Among them, several compounds have been tested at preclinical and clinical levels to treat glioblastoma multiforme (GBM). GBM is the most common and aggressive type of cancer originating in the brain and has an unfavorable prognosis, with a median survival of 15-16 months and a 5-year survival rate of 5%. Despite recent advances in treating GBM, it represents an incurable disease associated with treatment resistance and high recurrence rates. For these reasons, there is an urgent need for the development of new pharmacological agents to fight this malignancy. In this review, we reported the compounds published in the last five years, which showed promising activity in GBM preclinical models acting as TKIs. We grouped the compounds based on the targeted kinase: first, we reported receptor TKIs and then, cytoplasmic and peculiar kinase inhibitors. For each small molecule, we included the chemical structure, and we schematized the interaction with the target for some representative compounds with the aim of elucidating the mechanism of action. Finally, we cited the most relevant clinical trials.
Collapse
Affiliation(s)
| | | | | | - Francesca Musumeci
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (D.F.); (G.G.); (M.F.); (S.S.)
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (D.F.); (G.G.); (M.F.); (S.S.)
| | | |
Collapse
|
33
|
Singh S, Ghosh P, Roy R, Behera A, Sahadevan R, Kar P, Sadhukhan S, Sonawane A. 4″-Alkyl EGCG Derivatives Induce Cytoprotective Autophagy Response by Inhibiting EGFR in Glioblastoma Cells. ACS OMEGA 2024; 9:2286-2301. [PMID: 38250397 PMCID: PMC10795032 DOI: 10.1021/acsomega.3c06110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 01/23/2024]
Abstract
Epidermal growth factor receptor (EGFR)-targeted therapy has been proven vital in the last two decades for the treatment of multiple cancer types, including nonsmall cell lung cancer, glioblastoma, breast cancer and head and neck squamous cell carcinoma. Unfortunately, the majority of approved EGFR inhibitors fall into the drug resistance category because of continuous mutations and acquired resistance. Recently, autophagy has surfaced as one of the emerging underlying mechanisms behind resistance to EGFR-tyrosine kinase inhibitors (TKIs). Previously, we developed a series of 4″-alkyl EGCG (4″-Cn EGCG, n = 6, 8, 10, 12, 14, 16, and 18) derivatives with enhanced anticancer effects and stability. Therefore, the current study hypothesized that 4″-alkyl EGCG might induce cytoprotective autophagy upon EGFR inhibition, and inhibition of autophagy may lead to improved cytotoxicity. In this study, we have observed growth inhibition and caspase-3-dependent apoptosis in 4″-alkyl EGCG derivative-treated glioblastoma cells (U87-MG). We also confirmed that 4″-alkyl EGCG could inhibit EGFR in the cells, as well as mutant L858R/T790M EGFR, through an in vitro kinase assay. Furthermore, we have found that EGFR inhibition with 4″-alkyl EGCG induces cytoprotective autophagic responses, accompanied by the blockage of the AKT/mTOR signaling pathway. In addition, cytotoxicity caused by 4″-C10 EGCG, 4″-C12 EGCG, and 4″-C14 EGCG was significantly increased after the inhibition of autophagy by the pharmacological inhibitor chloroquine. These findings enhance our understanding of the autophagic response toward EGFR inhibitors in glioblastoma cells and suggest a potent combinatorial strategy to increase the therapeutic effectiveness of EGFR-TKIs.
Collapse
Affiliation(s)
- Satyam Singh
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Priya Ghosh
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Rajarshi Roy
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Ananyaashree Behera
- School
of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Orissa 751 024, India
| | - Revathy Sahadevan
- Department
of Chemistry, Indian Institute of Technology
Palakkad, Palakkad, Kerala 678 623, India
| | - Parimal Kar
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Sushabhan Sadhukhan
- Department
of Chemistry, Indian Institute of Technology
Palakkad, Palakkad, Kerala 678 623, India
| | - Avinash Sonawane
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| |
Collapse
|
34
|
Hoyt KW, Urul DA, Ogboo BC, Wittlinger F, Laufer SA, Schaefer EM, May EW, Heppner DE. Pitfalls and Considerations in Determining the Potency and Mutant Selectivity of Covalent Epidermal Growth Factor Receptor Inhibitors. J Med Chem 2024; 67:2-16. [PMID: 38134304 DOI: 10.1021/acs.jmedchem.3c01502] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Enzyme inhibitors that form covalent bonds with their targets are being increasingly pursued in drug development. Assessing their biochemical activity relies on time-dependent assays, which are distinct and more complex compared with methods commonly employed for reversible-binding inhibitors. To provide general guidance to the covalent inhibitor development community, we explored methods and reported kinetic values and experimental factors in determining the biochemical activity of various covalent epidermal growth factor receptor (EGFR) inhibitors. We showcase how liquid handling and assay reagents impact kinetic parameters and potency interpretations, which are critical for structure-kinetic relationships and covalent drug design. Additionally, we include benchmark kinetic values with reference inhibitors, which are imperative, as covalent EGFR inhibitor kinetic values are infrequently consistent in the literature. This overview seeks to inform best practices for developing new covalent inhibitors and highlight appropriate steps to address gaps in knowledge presently limiting assay reliability and reproducibility.
Collapse
Affiliation(s)
- Kristopher W Hoyt
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Daniel A Urul
- AssayQuant Technologies, Inc., Marlboro, Massachusetts 01752, United States
| | - Blessing C Ogboo
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Florian Wittlinger
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Stefan A Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - Erik M Schaefer
- AssayQuant Technologies, Inc., Marlboro, Massachusetts 01752, United States
| | - Earl W May
- AssayQuant Technologies, Inc., Marlboro, Massachusetts 01752, United States
| | - David E Heppner
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
- Department of Structural Biology, The State University of New York, Buffalo, New York 14203, United States
| |
Collapse
|
35
|
Park W, Han JH, Wei S, Yang ES, Cheon SY, Bae SJ, Ryu D, Chung HS, Ha KT. Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:807. [PMID: 38255882 PMCID: PMC10815680 DOI: 10.3390/ijms25020807] [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: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Targeted therapy against the epidermal growth factor receptor (EGFR) is a promising treatment approach for NSCLC. However, resistance to EGFR tyrosine kinase inhibitors (TKIs) remains a major challenge in its clinical management. EGFR mutation elevates the expression of hypoxia-inducible factor-1 alpha to upregulate the production of glycolytic enzymes, increasing glycolysis and tumor resistance. The inhibition of glycolysis can be a potential strategy for overcoming EGFR-TKI resistance and enhancing the effectiveness of EGFR-TKIs. In this review, we specifically explored the effectiveness of pyruvate dehydrogenase kinase inhibitors and lactate dehydrogenase A inhibitors in combating EGFR-TKI resistance. The aim was to summarize the effects of these natural products in preclinical NSCLC models to provide a comprehensive understanding of the potential therapeutic effects. The study findings suggest that natural products can be promising inhibitors of glycolytic enzymes for the treatment of EGFR-TKI-resistant NSCLC. Further investigations through preclinical and clinical studies are required to validate the efficacy of natural product-based glycolytic inhibitors as innovative therapeutic modalities for NSCLC.
Collapse
Affiliation(s)
- Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Shibo Wei
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Eun-Sun Yang
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea;
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Hwan-Suck Chung
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| |
Collapse
|
36
|
Shaban N, Kamashev D, Emelianova A, Buzdin A. Targeted Inhibitors of EGFR: Structure, Biology, Biomarkers, and Clinical Applications. Cells 2023; 13:47. [PMID: 38201251 PMCID: PMC10778338 DOI: 10.3390/cells13010047] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Members of the EGFR family of tyrosine kinase receptors are major regulators of cellular proliferation, differentiation, and survival. In humans, abnormal activation of EGFR is associated with the development and progression of many cancer types, which makes it an attractive target for molecular-guided therapy. Two classes of EGFR-targeted cancer therapeutics include monoclonal antibodies (mAbs), which bind to the extracellular domain of EGFR, and tyrosine kinase inhibitors (TKIs), which mostly target the intracellular part of EGFR and inhibit its activity in molecular signaling. While EGFR-specific mAbs and three generations of TKIs have demonstrated clinical efficacy in various settings, molecular evolution of tumors leads to apparent and sometimes inevitable resistance to current therapeutics, which highlights the need for deeper research in this field. Here, we tried to provide a comprehensive and systematic overview of the rationale, molecular mechanisms, and clinical significance of the current EGFR-targeting drugs, highlighting potential candidate molecules in development. We summarized the underlying mechanisms of resistance and available personalized predictive approaches that may lead to improved efficacy of EGFR-targeted therapies. We also discuss recent developments and the use of specific therapeutic strategies, such as multi-targeting agents and combination therapies, for overcoming cancer resistance to EGFR-specific drugs.
Collapse
Affiliation(s)
- Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Dmitri Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Aleksandra Emelianova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia;
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
37
|
Xi XX, Zhao HY, Mao YZ, Xin M, Zhang SQ. Modification of osimertinib to discover new potent EGFR C797S-TK inhibitors. Eur J Med Chem 2023; 261:115865. [PMID: 37839342 DOI: 10.1016/j.ejmech.2023.115865] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
The EGFRC797S mutation is a dominant mechanism of acquired resistance after the treatment of non-small cell lung cancer (NSCLC) with osimertinib in clinic. To date, there is no inhibitor approved to overcome the resistance caused by osimertinib. In this study, a series of compounds with phenylamino-pyrimidine scaffold deriving from osimertinib were designed, synthesized and evaluated as fourth-generation EGFRC797S-TK inhibitors. Consequently, compound Os30 exhibited potent inhibitory activities against both EGFRDel19/T790M/C797S TK and EGFRL858R/T790M/C797S TK with IC50 values of 18 nM and 113 nM, respectively. Moreover, Os30 can powerfully inhibit the proliferation of KC-0116 (BaF3-EGFRDel19/T790M/C797S) and KC-0122 (BaF3-EGFRL858R/T790M/C797S) cells. In addition, Os30 can suppress EGFR phosphorylation in a concentration-dependent manner in KC-0116 cells, arrest KC-0116 cells at G1 phase and induce the apoptosis of KC-0116 cells. More importantly, Os30 showed potent antitumor efficacy in the KC-0116 cells xenograft nude mice tumor model with the tumor growth inhibitory rate of 77.6% at a dosage of 40 mg/kg. These findings demonstrate that modification of osimertinib can discover new potent EGFRC797S-TK inhibitors, and compound Os30 is a potent fourth-generation EGFR inhibitor to treat NSCLC with EGFmRC797S mutation.
Collapse
Affiliation(s)
- Xiao-Xiao Xi
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Hong-Yi Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yu-Ze Mao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
38
|
Hou G, Niu T, Jia A, Zhang Y, Chen X, Wei H, Jia Y, Xu Y, Li Y, Wang P, Chatterjee A. NRG1 promotes tumorigenesis and metastasis and afatinib treatment efficiency is enhanced by NRG1 inhibition in esophageal squamous cell carcinoma. Biochem Pharmacol 2023; 218:115920. [PMID: 37989416 DOI: 10.1016/j.bcp.2023.115920] [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: 10/02/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly aggressive tumor with significant heterogeneity in incidence and outcomes. The role of Neuregulin 1 (NRG1) in ESCC and its contribution to aggressiveness remain unknown. This study aims to investigate the functions and molecular mechanisms of NRG1 in ESCC as well as the treatment strategy for ESCC with overexpression of NRG1. We firstly demonstrated the upregulation of NRG1 and a negative correlation trend between patients' overall survival (OS) and the expression level of NRG1 in esophageal cancer. And then we found NRG1 promoted cell proliferation, migration, inhibited apoptosis, and accelerated tumorigenesis and metastasis in ESCC using cell lines and xenograft models. Furthermore, we discovered that NRG1 activated the NF-κB/MMP9 signaling pathway, contributing to the metastatic phenotype in ESCC. Finally, we show that afatinib (FDA approved cancer growth blocker) could inhibit ESCC with overexpressed NRG1 and down-regulation of NRG1 along with afatinib treatment provides higher efficient strategy. This study uncovers the critical role and molecular mechanism of NRG1 in ESCC tumorigenesis and metastasis, suggesting its potential as a novel biomarker for ESCC treatment.
Collapse
Affiliation(s)
- Guiqin Hou
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Tengda Niu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ang Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yingying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xunan Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Huiyun Wei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yilin Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yichao Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Li
- Center of Advanced Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou 450001, China
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; School of Health Sciences and Technology, UPES, Dehradun, India.
| |
Collapse
|
39
|
Zhou S, Zhao FL, Wang SH, Wang YR, Hong Y, Zhou Q, Geng PW, Luo QF, Cai JP, Dai DP. Assessments of CYP‑inhibition‑based drug-drug interaction between vonoprazan and poziotinib in vitro and in vivo. PHARMACEUTICAL BIOLOGY 2023; 61:356-361. [PMID: 36728978 PMCID: PMC9897767 DOI: 10.1080/13880209.2023.2173253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/16/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Poziotinib and vonoprazan are two drugs mainly metabolized by CYP3A4. However, the drug-drug interaction between them is unknown. OBJECTIVE To study the interaction mechanism and pharmacokinetics of poziotinib on vonoprazan. MATERIALS AND METHODS In vitro experiments were performed with rat liver microsomes (RLMs) and the contents of vonoprazan and its metabolite were then determined with UPLC-MS/MS after incubation of RLMs with vonoprazan and gradient concentrations of poziotinib. For the in vivo experiment, rats in the poziotinib treated group were given 5 mg/kg poziotinib by gavage once daily for 7 days, and the control group was only given 0.5% CMC-Na. On Day 8, tail venous blood was collected at different time points after the gavage administration of 10 mg/kg vonoprazan, and used for the quantification of vonoprazan and its metabolite. DAS and SPSS software were used for the pharmacokinetic and statistical analyses. RESULTS In vitro experimental data indicated that poziotinib inhibited the metabolism of vonoprazan (IC50 = 10.6 μM) in a mixed model of noncompetitive and uncompetitive inhibition. The inhibitory constant Ki was 0.574 μM and the binding constant αKi was 2.77 μM. In vivo experiments revealed that the AUC(0-T) (15.05 vs. 90.95 μg/mL·h) and AUC(0-∞) (15.05 vs. 91.99 μg/mL·h) of vonoprazan increased significantly with poziotinib pretreatment. The MRT(0-∞) of vonoprazan increased from 2.29 to 5.51 h, while the CLz/F value decreased from 162.67 to 25.84 L/kg·h after pretreatment with poziotinib. CONCLUSIONS Poziotinib could significantly inhibit the metabolism of vonoprazan and more care may be taken when co-administered in the clinic.
Collapse
Affiliation(s)
- Shan Zhou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Fang-Ling Zhao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
- Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Shuang-Hu Wang
- Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Yi-Ran Wang
- Peking University Fifth School of Clinical Medicine, Beijing, China
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Yun Hong
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Quan Zhou
- Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Pei-Wu Geng
- Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Qing-Feng Luo
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Da-Peng Dai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
- Peking University Fifth School of Clinical Medicine, Beijing, China
| |
Collapse
|
40
|
Gómez-Oliva R, Geribaldi-Doldán N, Domínguez-García S, Pardillo-Díaz R, Martínez-Ortega S, Oliva-Montero JM, Pérez-García P, García-Cózar FJ, Muñoz-Miranda JP, Sánchez-Gomar I, Nunez-Abades P, Castro C. Targeting epidermal growth factor receptor to recruit newly generated neuroblasts in cortical brain injuries. J Transl Med 2023; 21:867. [PMID: 38037126 PMCID: PMC10687845 DOI: 10.1186/s12967-023-04707-1] [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: 02/03/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Neurogenesis is stimulated in the subventricular zone (SVZ) of mice with cortical brain injuries. In most of these injuries, newly generated neuroblasts attempt to migrate toward the injury, accumulating within the corpus callosum not reaching the perilesional area. METHODS We use a murine model of mechanical cortical brain injury, in which we perform unilateral cortical injuries in the primary motor cortex of adult male mice. We study neurogenesis in the SVZ and perilesional area at 7 and 14 dpi as well as the expression and concentration of the signaling molecule transforming growth factor alpha (TGF-α) and its receptor the epidermal growth factor (EGFR). We use the EGFR inhibitor Afatinib to promote neurogenesis in brain injuries. RESULTS We show that microglial cells that emerge within the injured area and the SVZ in response to the injury express high levels of TGF-α leading to elevated concentrations of TGF-α in the cerebrospinal fluid. Thus, the number of neuroblasts in the SVZ increases in response to the injury, a large number of these neuroblasts remain immature and proliferate expressing the epidermal growth factor receptor (EGFR) and the proliferation marker Ki67. Restraining TGF-α release with a classical protein kinase C inhibitor reduces the number of these proliferative EGFR+ immature neuroblasts in the SVZ. In accordance, the inhibition of the TGF-α receptor, EGFR promotes migration of neuroblasts toward the injury leading to an elevated number of neuroblasts within the perilesional area. CONCLUSIONS Our results indicate that in response to an injury, microglial cells activated within the injury and the SVZ release TGF-α, activating the EGFR present in the neuroblasts membrane inducing their proliferation, delaying maturation and negatively regulating migration. The inactivation of this signaling pathway stimulates neuroblast migration toward the injury and enhances the quantity of neuroblasts within the injured area. These results suggest that these proteins may be used as target molecules to regenerate brain injuries.
Collapse
Affiliation(s)
- Ricardo Gómez-Oliva
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Noelia Geribaldi-Doldán
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Ricardo Pardillo-Díaz
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Sergio Martínez-Ortega
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - José M Oliva-Montero
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Patricia Pérez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Francisco J García-Cózar
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Área de Inmunología, Universidad de Cádiz, Cádiz, Spain
| | - Juan P Muñoz-Miranda
- Servicios Centrales de Investigación Biomédica, Universidad de Cádiz, Cádiz, Spain
| | - Ismael Sánchez-Gomar
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Departamento de Fisiología, Universidad de Sevilla, Sevilla, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain.
| |
Collapse
|
41
|
Duan ZK, Guo SS, Ye L, Gao ZH, Liu D, Yao GD, Song SJ, Huang XX. Discovery of Michael reaction acceptors from the leaves of Ailanthus altissima by a modified tactic. PHYTOCHEMISTRY 2023; 215:113858. [PMID: 37709157 DOI: 10.1016/j.phytochem.2023.113858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/09/2023] [Accepted: 09/09/2023] [Indexed: 09/16/2023]
Abstract
Structural characteristics-guided investigation of Ailanthus altissima (Mill.) Swingle resulted in the isolation and identification of seven undescribed potential Michael reaction acceptors (1-7). Ailanlactone A (1) possesses an unusual 1,7-epoxy-11,12-seco quassinoid core. Ailanterpene B (6) was a rare guaianolide-type sesquiterpene with a 5/6/6/6-fused skeleton. Their structures were determined through extensive analysis of physiochemical and spectroscopic data, quantum chemical calculations, and single crystal X-ray crystallographic technology using Cu Kα radiation. The cytotoxic activities of isolates on HepG2 and Hep3B cells were evaluated in vitro. Encouragingly, ailanaltiolide K (4) showed significant cytotoxicity against Hep3B cells with IC50 values of 1.41 ± 0.21 μM, whose covalent binding mode was uncovered in silico.
Collapse
Affiliation(s)
- Zhi-Kang Duan
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shan-Shan Guo
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Li Ye
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhi-Heng Gao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Dai Liu
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiao-Xiao Huang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
42
|
Othman DI, Hamdi A, Elhusseiny WM, El-Azab AS, Bakheit AH, Hefnawy M, Abdel-Aziz AAM. Synthesis of novel spirochromane incorporating Schiff's bases, potential antiproliferative activity, and dual EGFR/HER2 inhibition: Cell cycle analysis and in silico study. Saudi Pharm J 2023; 31:101803. [PMID: 37860686 PMCID: PMC10582582 DOI: 10.1016/j.jsps.2023.101803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
Spirochromanes incorporating Schiff's bases and semicarbazones 4a-e and 5a-j were synthesizedand analyzed for their potential antiproliferative activity using four human cancer cell lines (MCF-7, HCT-116, PC3, and A549). Compounds 5a, 5b and 5g possessed the highest antiproliferative activity among the tested compounds,with an IC50 range of 1.154-9.09 μM. Compound 5j selectively inhibited the PC3 cell proliferation (IC50 = 5.47 μM). Spirochromanes 5a, 5b and 5g exhibited high inhibitory activity against EGFR (IC50 = 0.116, 0.132, and 0.077 μM, respectively) and HER2 (IC50 = 0.055, 0.210 and 0.085 μM, respectively) compared with the references, erlotinib (IC50 = 0.090 and 0.038 μM, respectively) and gefitinib (IC50 = 0.052 and 0.072 μM, respectively). Cell cycle analysis and apoptosis results showed that compounds 5a, 5b and 5g arrested growth inthe S phase, and the programmed cell death induced by these compounds was an apoptotic mechanism rather than a necrotic pathway. Molecular docking studies of spirochromanes 5a, 5b and 5g to EGFR and HER2 binding sites were performed to explore the orientation mode and interaction.
Collapse
Affiliation(s)
- Dina I.A. Othman
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Walaa M. Elhusseiny
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Adel S. El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed H. Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alaa A.-M. Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
43
|
Peng J, Li P, Li Y, Quan J, Yao Y, Duan J, Liu X, Li H, Yuan D, Wang X. PFKP is a prospective prognostic, diagnostic, immunological and drug sensitivity predictor across pan-cancer. Sci Rep 2023; 13:17399. [PMID: 37833332 PMCID: PMC10576092 DOI: 10.1038/s41598-023-43982-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023] Open
Abstract
Phosphofructokinase, platelet (PFKP) is a rate-limiting enzyme of glycolysis that plays a decisive role in various human physio-pathological processes. PFKP has been reported to have multiple functions in different cancer types, including lung cancer and breast cancer. However, no systematic pancancer analysis of PFKP has been performed; this type of analysis could elucidate the clinical value of PFKP in terms of diagnosis, prognosis, drug sensitivity, and immunological correlation. Systematic bioinformation analysis of PFKP was performed based on several public datasets, including The Cancer Genome Atlas (TCGA), Cancer Cell Line Encyclopedia (CCLE), Genotype-Tissue Expression Project (GTEx), and Human Protein Atlas (HPA). Prospective carcinogenesis of PFKP across cancers was estimated by expression analysis, effect on patient prognosis, diagnosis significance evaluation, and immunity regulation estimation. Then, pancancer functional enrichment of PFKP was also assessed through its effect on the signaling score and gene expression profile. Finally, upstream expression regulation of PFKP was explored by promoter DNA methylation and transcription factor (TF) prediction. Our analysis revealed that high expression of PFKP was found in most cancer types. Additionally, a high level of PFKP displayed a significant correlation with poor prognosis in patients across cancers. The diagnostic value of PFKP was performed based on its positive correlation with programmed cell death-ligand 1 (PD-L1). We also found an obvious immune-regulating effect of PFKP in most cancer types. PFKP also had a strong negative correlation with several cancer drugs. Finally, ectopic expression of PFKP may depend on DNA methylation and several predicated transcription factors, including the KLF (KLF transcription factor) and Sp (Sp transcription factor) families. This pancancer analysis revealed that a high expression level of PFKP might be a useful biomarker and predictor in most cancer types. Additionally, the performance of PFKP across cancers also suggested its meaningful role in cancer immunity regulation, even in immunotherapy and drug resistance. Overall, PFKP might be explored as an auxiliary monitor for pancancer early prognosis and diagnosis.
Collapse
Affiliation(s)
- Jian Peng
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Pingping Li
- Comprehensive Liver Cancer Center, The Fifth Medical Center of the PLA General Hospital, Beijing, 100039, China
| | - Yuan Li
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jichuan Quan
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100020, China
| | - Yanwei Yao
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junfang Duan
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xuemei Liu
- Department of Respiratory and Critical Care Medicine, Second People's Hospital of Taiyuan, Taiyuan, 030002, Shanxi, China
| | - Hao Li
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Dajiang Yuan
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Xiaoru Wang
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
44
|
He H, Liu JQ, Wang XS. Synthesis of pyrimidine-fused skeletons through copper-catalyzed consecutive Sonogashira coupling and aminocyclization. Org Biomol Chem 2023; 21:7886-7890. [PMID: 37743708 DOI: 10.1039/d3ob01248d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
A copper-catalyzed sequence involving a Sonogashira coupling reaction and 5-exo-dig aminocyclization between a terminal alkyne and a 2-(2-bromophenyl)pyrimidine analog based on six-membered rings is presented. This protocol provides a controlled and modular approach to access a variety of synthetically useful pyrimidine-fused skeletons with high efficiency, broad substrate scope, and excellent functional group compatibility. Under acidic conditions, the (Z)-configuration of products spontaneously converts into (E)-9-benzylidene-1,9-dihydro-11H-pyrazolo[4',3':4,5]pyrimido[2,1-a]isoindol-11-ones.
Collapse
Affiliation(s)
- Heng He
- School of Chemistry and Materials Science, Jiangsu Key Laboratory of Green Synthesis for Functional Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China.
| | - Jian-Quan Liu
- School of Chemistry and Materials Science, Jiangsu Key Laboratory of Green Synthesis for Functional Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China.
| | - Xiang-Shan Wang
- School of Chemistry and Materials Science, Jiangsu Key Laboratory of Green Synthesis for Functional Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China.
| |
Collapse
|
45
|
Mortazavi M, Eskandari M, Moosavi F, Damghani T, Khoshneviszadeh M, Pirhadi S, Saso L, Edraki N, Firuzi O. Novel quinazoline-1,2,3-triazole hybrids with anticancer and MET kinase targeting properties. Sci Rep 2023; 13:14685. [PMID: 37673888 PMCID: PMC10482942 DOI: 10.1038/s41598-023-41283-2] [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: 03/06/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
Oncogenic activation of receptor tyrosine kinases (RTKs) such as MET is associated with cancer initiation and progression. We designed and synthesized a new series of quinazoline derivatives bearing 1,2,3-triazole moiety as targeted anticancer agents. The MET inhibitory effect of synthesized compounds was assessed by homogeneous time-resolved fluorescence (HTRF) assay and western blot analysis. Sulforhodamine B assay was conducted to examine the antiproliferative effects of synthetic compounds against 6 cancer cell lines from different origins including MET-dependent AsPC-1, EBC-1 and MKN-45 cells and also Mia-Paca-2, HT-29 and K562 cells. The growth inhibitory effect of compounds in a three-dimensional spheroid culture was examined by acid phosphatase (APH) assay, while apoptosis induction was evaluated by Annexin V/propidium iodide method. Compound 8c bearing p-methyl benzyl moiety on the triazole ring exhibited the highest MET inhibitory capacity among tested agents that was further confirmed by western blot findings. Derivatives 8c and 8h exhibited considerable antiproliferative effects against all tested cell lines, with more inhibitory effects against MET-positive cells with IC50 values as low as 6.1 μM. These two agents also significantly suppressed cell growth in spheroid cultures and induced apoptosis in MET overexpressing AsPC-1 cells. Moreover, among a panel of 24 major oncogenic kinases, the PDGFRA kinase was identified as a target of 8c and 8h compounds. The docking study results of compounds 8c and 8h were in agreement with experimental findings. The results of the present study suggest that quinazoline derivatives bearing 1,2,3-triazole moiety may represent promising targeted anticancer agents.
Collapse
Affiliation(s)
- Motahareh Mortazavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoomeh Eskandari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Damghani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Pirhadi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
46
|
Li P, Li B, Yang N, Xu T, Zheng Z. The next generation of EGFR inhibitors: a patenting perspective of PROTACs based EGFR degraders. Expert Opin Ther Pat 2023; 33:477-492. [PMID: 37873645 DOI: 10.1080/13543776.2023.2262176] [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: 05/22/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION Abnormal expression of epidermal growth factor receptor (EGFR) contributes to tumor development, especially in non-small cell lung cancer (NSCLC). Although multiple inhibitors have been developed to target diverse EGFR mutations and several have been approved, the inevitable drug resistance and side effect remain a challenge, which motivates novel strategies. Proteolysis-targeting chimeras (PROTACs) have been gaining momentum for their potential as novel therapeutics for human diseases by triggering protein degradation. To date, various potent and specific EGFR PROTACs have been discovered and some of them have entered clinical trials. AREAS COVERED This review provides an overview of EGFR degraders in patents from 2016 to 2022. It provides an update of the discovery strategies, chemical structures, and molecular profiling of all available EGFR PROTACs. SciFinder, PubMed, Web of Science, EPO, and CNIPA databases were used for searching the literature and patents for EGFR PROTACs. EXPERT OPINION By employing the PROTAC technology, highly potent and selective EGFR degraders based on four generation EGFR inhibitors have been developed, which offer a new strategy to target EGFR mutations and overcome the drug resistance. Despite the satisfactory result in vitro and in vivo studies, their therapeutic value awaits more rigorous preclinical testing and clinical investigation.
Collapse
Affiliation(s)
- Pengyun Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Bingkun Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ning Yang
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Tingting Xu
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhibing Zheng
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
47
|
Godesi S, Nada H, Lee J, Kang JH, Kim SY, Choi Y, Lee K. Integration of Hybridization Strategies in Pyridine-Urea Scaffolds for Novel Anticancer Agents: Design, Synthesis, and Mechanistic Insights. Molecules 2023; 28:4952. [PMID: 37446614 DOI: 10.3390/molecules28134952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Annually, millions of new cancer cases are reported, leading to millions of deaths worldwide. Among the newly reported cases, breast and colon cancers prevail as the most frequently detected variations. To effectively counteract this rapid increase, the development of innovative therapies is crucial. Small molecules possessing pyridine and urea moieties have been reported in many of the currently available anticancer agents, especially VEGFR2 inhibitors. With this in mind, a rational design approach was employed to create hybrid small molecules combining urea and pyridine. These synthesized compounds underwent in vitro testing against breast and colon cancer cell lines, revealing potent submicromolar anticancer activity. Compound 8a, specifically, exhibited an impressive GI50 value of 0.06 μM against the MCF7 cancer cell line, while compound 8h displayed the highest cytotoxic activity against the HCT116 cell line, with a GI50 of 0.33 ± 0.042 μM. Notably, compounds 8a, 8h, and 8i demonstrated excellent safety profiles when tested on normal cells. Molecular docking, dynamic studies, and free energy calculations were employed to validate the affinity of these compounds as VEGFR2 inhibitors.
Collapse
Affiliation(s)
- Sreenivasulu Godesi
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Hossam Nada
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Cairo, Cairo 11829, Egypt
| | - Joohan Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Joon-Hee Kang
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Soo-Youl Kim
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Yongseok Choi
- College of Biosciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Kyeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| |
Collapse
|
48
|
Guo Y, Gao B, Gao P, Wang Y, Gou S. Design, synthesis and biological evaluation of phosphoroxy quinazoline derivatives as potential EGFR T790M/C797S inhibitors. Bioorg Med Chem 2023; 90:117338. [PMID: 37269687 DOI: 10.1016/j.bmc.2023.117338] [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: 04/10/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 06/05/2023]
Abstract
Owing to the urgency and importance of developing fourth-generation EGFR inhibitors that can effectively overcome C797S site mutation in NSCLC, Brigatinib was used in this work as a lead compound to modify its structure to obtain a series of phosphoroxy quinazoline derivatives. Biological study indicated that the inhibitory activity and selectivity of the target compounds on EGFRL858R/T790M/C797S/EGFRDel19/T790M/C797S enzymes and EGFRDel19/T790M/C797S overexpressed Ba/F3 cells were significantly better than those of Brigatinib. Among the target compounds, 8a exhibited the best biological activity in vitro. More importantly, 8a presented acceptable pharmacokinetic behaviors and showed potent anti-tumor efficacy in the Ba/F3-EGFRDel19/T790M/C797S subcutaneous xenograft mice model with the tumor growth inhibition value of 82.60% at a dose of 30 mg/kg. These results indicated that 8a, as a drug candidate of the novel fourth-generation EGFR small-molecule inhibitor, has high potentials to treat with NSCLC on EGFR with C797S mutation.
Collapse
Affiliation(s)
- Yanliang Guo
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China; Jiangsu Hansoh Pharmaceutical Group CO., LTD., Lianyungang 222000, PR China
| | - Biao Gao
- Jiangsu Hansoh Pharmaceutical Group CO., LTD., Lianyungang 222000, PR China
| | - Peng Gao
- Jiangsu Hansoh Pharmaceutical Group CO., LTD., Lianyungang 222000, PR China
| | - Yuanjiang Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China.
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
49
|
Gollner A, Heine C, Hofbauer KS. Kinase Degraders, Activators, and Inhibitors: Highlights and Synthesis Routes to the Chemical Probes on opnMe.com, Part 1. ChemMedChem 2023; 18:e202300031. [PMID: 36825440 DOI: 10.1002/cmdc.202300031] [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: 01/20/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
Kinases are among the most important and successful drug targets. Chemical probe compounds have played a critical role in elucidating the role of kinases in many biological pathways. There are currently twelve well-validated chemical probes that target kinases available free-of-cost via the Molecules to Order (M2O) arm of Boehringer Ingelheim's open innovation platform, opnMe.com. Here we present a summary of the key data for each of these probe compounds and the synthesis routes to all twelve compounds. We hope this will aid researchers who use or plan to use these compounds in their research.
Collapse
Affiliation(s)
- Andreas Gollner
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co. KG, Boehringer-Gasse, Wien, 5-11, 1121 Vienna, Austria
| | - Claudia Heine
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co. KG, 88400, Biberach, Germany
| | - Karin S Hofbauer
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co. KG, Boehringer-Gasse, Wien, 5-11, 1121 Vienna, Austria
| |
Collapse
|
50
|
Schaefer D, Cheng X. Recent Advances in Covalent Drug Discovery. Pharmaceuticals (Basel) 2023; 16:ph16050663. [PMID: 37242447 DOI: 10.3390/ph16050663] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/10/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
In spite of the increasing number of biologics license applications, the development of covalent inhibitors is still a growing field within drug discovery. The successful approval of some covalent protein kinase inhibitors, such as ibrutinib (BTK covalent inhibitor) and dacomitinib (EGFR covalent inhibitor), and the very recent discovery of covalent inhibitors for viral proteases, such as boceprevir, narlaprevir, and nirmatrelvir, represent a new milestone in covalent drug development. Generally, the formation of covalent bonds that target proteins can offer drugs diverse advantages in terms of target selectivity, drug resistance, and administration concentration. The most important factor for covalent inhibitors is the electrophile (warhead), which dictates selectivity, reactivity, and the type of protein binding (i.e., reversible or irreversible) and can be modified/optimized through rational designs. Furthermore, covalent inhibitors are becoming more and more common in proteolysis, targeting chimeras (PROTACs) for degrading proteins, including those that are currently considered to be 'undruggable'. The aim of this review is to highlight the current state of covalent inhibitor development, including a short historical overview and some examples of applications of PROTAC technologies and treatment of the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Daniel Schaefer
- Buchmann Institute for Molecular Life Sciences, Chemical Biology, Goethe University Frankfurt am Main, Max-von-Laue-Strasse 15. R. 3.652, 60438 Frankfurt am Main, Germany
- Pharmaceutical Chemistry, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Xinlai Cheng
- Buchmann Institute for Molecular Life Sciences, Chemical Biology, Goethe University Frankfurt am Main, Max-von-Laue-Strasse 15. R. 3.652, 60438 Frankfurt am Main, Germany
- Pharmaceutical Chemistry, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|