1
|
Fan Y, Liu Y, Ouyang X, Su J, Zhou X, Jia Q, Chen W, Chen W, Liu X. Prediction of EGFR mutation status and its subtypes in non-small cell lung cancer based on 18F-FDG PET/CT radiological features. Nucl Med Commun 2025:00006231-990000000-00386. [PMID: 39829249 DOI: 10.1097/mnm.0000000000001948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
PURPOSE Prediction of epidermal growth factor receptor (EGFR) mutation status and subtypes in patients with non-small cell lung cancer (NSCLC) based on 18F-fluorodeoxyglucose (18F-FDG) PET/computed tomography (CT) radiomics features. PATIENTS AND METHODS Retrospective analysis of 201 NSCLC patients with 18F-FDG PET/CT and EGFR genetic testing was carried out. Radiomics features and clinical factors were used to construct a combined model for identifying EGFR mutation status. Mutation/wild-type models were trained in a training cohort (n = 129) and validated in an internal validation cohort (n = 41) vs an external validation cohort (n = 50). A second model predicting the 19/21 mutation locus was also built and evaluated in a subset of EGFR mutations (training cohort, n = 55; validation cohort, n = 14). The predictive performance and net clinical benefit of the models were assessed by analysis of the area under curve (AUC) of the subjects, nomogram, calibration curve and decision curve. RESULTS The AUC of the combined model distinguishing EGFR mutation status was 0.864 in the training cohort and 0.806 and 0.791 in the internal vs external test sets respectively, and the AUC of the 19/21 mutation site model was 0.971 and 0.867 in the training cohort and internal validation cohort respectively. The calibration curves of the individual models showed better model predictions (Brier score <0.25). Decision curve analysis showed that the models had clinical application. CONCLUSION The combined model based on 18F-FDG PET/CT radiomics features combined and clinical features can predict EGFR mutation status and subtypes in NSCLC patients, and guiding targeted therapy, and facilitate precision medicine development.
Collapse
Affiliation(s)
- Yishuo Fan
- Department of Graduate School, Graduate School of Hebei North University, Zhangjiakou, Hebei
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| | - Yuang Liu
- Department of Graduate School, Graduate School of Hebei North University, Zhangjiakou, Hebei
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| | - Xiaohui Ouyang
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| | - Jiagui Su
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| | - Xiaohong Zhou
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| | - Qichen Jia
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| | - Wenjing Chen
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaofei Liu
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital
| |
Collapse
|
2
|
An Y, Lv X, Xu S, Li H, Zheng P, Zhu W, Wang L. Pyrimidine-based dual-target inhibitors targeting epidermal growth factor receptor for overcoming drug resistance in cancer therapy(2006-present). Eur J Med Chem 2025; 286:117268. [PMID: 39837171 DOI: 10.1016/j.ejmech.2025.117268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
The epidermal growth factor receptor (EGFR) is a pivotal member of the epidermal growth factor receptor family, exerting crucial regulatory influence on cellular physiological processes, particularly in relation to cell growth, proliferation, and differentiation. In recent years, numerous EGFR inhibitors have been introduced to the market; unfortunately, the effectiveness of single-target EGFR inhibitors has been compromised due to the development of drug resistance caused by EGFR mutations. Despite attempts by some researchers to address this issue through combination therapy with two or more drugs, instances of dose-limiting toxicities have been observed. Consequently, EGFR dual-target inhibitors have emerged as a burgeoning field in cancer treatment, offering a novel therapeutic option for solid tumors with the added benefits of reduced risk of resistance, lower dosage requirements, diminished toxicity profiles, and enhanced efficacy. At present, a series of EGFR dual-target inhibitors with diverse structures have been developed successively. In this study, we initially investigated the pyrimidine-based EGFR dual-target inhibitors that have been reported in the past two decades and categorized them into aminopyrimidine derivatives and heterocyclic pyrimidine derivatives with increased molecular complexity. Subsequently, we comprehensively summarized the biological activity and structure-activity relationship of this class of inhibitors in the context of cancer therapy, while also exploring potential opportunities and challenges associated with their application in this field. The present study provides a partial framework to guide future endeavors in drug development.
Collapse
Affiliation(s)
- Yufeng An
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Xinya Lv
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Shidi Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Heqing Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| | - Linxiao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| |
Collapse
|
3
|
Wei G, Nie Y, Sun M, Zhou W, Zhao H, Chen F, Zhu C. Cancer-associated fibroblasts induce almonertinib resistance in non-small cell lung cancer. J Transl Med 2025; 23:42. [PMID: 39794783 PMCID: PMC11724582 DOI: 10.1186/s12967-024-06064-z] [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: 08/14/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Almonertinib is the initial third-generation EGFR-TKI in China, but its resistance mechanism is unknown. Cancer-associated fibroblasts (CAFs) are essential matrix components in the tumor microenvironment, but their impact on almonertinib resistance is unknown. This study aimed to explore the correlation between CAFs and almonertinib resistance in non-small cell lung cancer (NSCLC). METHODS The anti-cancer effects of almonertinib on NSCLC cells, as well as the reversal of these effects mediated by CAFs, were validated through phenotypic experiments. Differential gene expression analysis, along with GO and KEGG enrichment analyses, was performed to predict the potential mechanisms underlying resistance to third-generation EGFR-TKIs. Finally, qPCR and Western blot analyses were used to explore the signaling pathways by which CAFs induce resistance to almonertinib in NSCLC cells. RESULTS Our findings revealed that almonertinib significantly suppressed the invasion, migration, and proliferation of EGFR T790M-mutant NSCLC cells. TGF-β1 successfully induced the differentiation of CAFs and upregulated the expression of CAF markers, including α-SMA and fibroblast activation protein (FAP). Exposure of H1975 cells to almonertinib increased TGF-β1 secretion. Additionally, CAFs enhanced the survival of almonertinib-treated NSCLC cells, whereas normal fibroblasts (NFs) exerted the opposite effect. qPCR analysis demonstrated that the expression of the core molecules of the Hippo pathway, YAP and TAZ, was lower in A549 cells than in H1975 cells, and CAF intervention further reduced YAP/TAZ expression in H1975 cells. Western blot analysis confirmed a significant reduction in YAP/TAZ protein levels in cancer cells treated with CAF-conditioned medium (CAF-CM) compared to those treated with normal control-conditioned medium (NC-CM). Finally, we demonstrated that CAFs induced resistance to almonertinib in NSCLC cells, potentially through a mechanism involving YAP/TAZ. CONCLUSION This study demonstrated that H1975 cells stimulated by almonertinib promoted the accumulation of CAFs in NSCLC cells, likely through increased secretion of TGF-β1. The accumulation of CAFs enhanced the survival of NSCLC cells undergoing almonertinib treatment and induced drug resistance. Additionally, the mechanism underlying CAF-induced drug resistance in NSCLC cells was potentially linked to the activation of the YAP/TAZ signaling pathway.
Collapse
Affiliation(s)
- Guohao Wei
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Yu Nie
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Min Sun
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Wenzheng Zhou
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Huihui Zhao
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
- Department of Oncology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| | - Fangfang Chen
- Department of Clinical Laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Chuandong Zhu
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| |
Collapse
|
4
|
Sun K, Wang P. A good response to furmonertinib fourth-line treatment of an advanced lung adenocarcinoma patient with EGFR exon20in and PIK3CA mutation: a case report and literature review. Front Oncol 2024; 14:1467722. [PMID: 39743996 PMCID: PMC11688600 DOI: 10.3389/fonc.2024.1467722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Background Lung cancer, including small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), is the most prevalent cancer globally and remains the leading cause of cancer-related mortality. Epidermal growth factor receptor (EGFR) mutations, frequently observed in female NSCLC patients, have revolutionized treatment strategies with the advent of tyrosine kinase inhibitors (TKIs). These therapies significantly improve survival and are considered the standard of care for patients harboring EGFR mutations. However, most patients eventually develop resistance to EGFR-TKIs, leading to disease progression. Resistance mechanisms are classified as either EGFR-dependent or EGFR-independent, the latter involving bypass pathway activation, including dysregulation of downstream signaling cascades. EGFR-independent resistance often renders all EGFR-TKIs ineffective, necessitating further investigation into resistance mechanisms. Case summary We report the case of a 63-year-old Chinese woman diagnosed with synchronous lung adenocarcinoma harboring an EGFR exon 21 far-loop insertion mutation and clear cell renal cell carcinoma (ccRCC). A multidisciplinary team recommended systemic therapy for the lung adenocarcinoma and clinical observation for ccRCC. First-line treatment with bevacizumab plus pemetrexed-carboplatin achieved a progression-free survival (PFS) of 7 months. Second-line treatment with sintilimab and nedaplatin resulted in a PFS of 4.9 months. Third-line therapy with sintilimab and anlotinib proved ineffective. In the fourth line, the patient received furmonertinib, a third-generation EGFR-TKI, based on the FAVOUR trial. This treatment achieved durable disease control with excellent tolerability, yielding a PFS of 27 months and ongoing clinical benefit. Conclusion This case demonstrates that furmonertinib can provide significant clinical benefit to NSCLC patients with complex resistance mechanisms, including those involving the PIK3CA/mTOR pathway. These findings support its potential to overcome EGFR-TKI resistance and warrant further investigation in similar clinical contexts.
Collapse
Affiliation(s)
| | - Peng Wang
- Department Oncology, Yidu Central Hospital of Weifang, Weifang, China
| |
Collapse
|
5
|
Li KM, Meng LF, Yang ZH, Hu WT. NUP155 and NDC1 interaction in NSCLC: a promising target for tumor progression. Front Pharmacol 2024; 15:1514367. [PMID: 39720592 PMCID: PMC11666513 DOI: 10.3389/fphar.2024.1514367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 11/28/2024] [Indexed: 12/26/2024] Open
Abstract
Background NUP155 was reported to involve breast invasive carcinoma and hepatocellular carcinoma. We hypothesized that NUP155 and NDC1impacted the progression of NSCLC. Methods The dataset was analyzed to find differentially expressed genes. Functional enrichment analysis and Kaplan-Meier survival analysis were performed for differentially expressed genes. Western blot, Clone formation assay, Transwell assay and CCK-8 assay were performed to determine the performance and role of the target gene in NSCLC. Results The research found that the NUP family played a role in various diseases. Differential expression analysis and survival analysis were performed to identify 6 related-genes, including NUP155, NDC1, KPNA2, MAD2L1, NUP62CL, and POM121L2NUP155 and NDC1 could interact with NUP53, respectively. This effect was necessary to complete the assembly of the nuclear pore complex. Conclusion NUP155 interacted with NDC1 to complete the assembly of the nuclear pore complex, which promoted the development of NSCLC. Our study demonstrated that NUP155 was expected to be a potential target for the treatment of NSCLC.
Collapse
Affiliation(s)
| | | | | | - Wen-Tao Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
6
|
Wang Z, Xie S, Li L, Liu Z, Zhou W. Schisandrin C inhibits AKT1-regulated cell proliferation in A549 cells. Int Immunopharmacol 2024; 142:113110. [PMID: 39260306 DOI: 10.1016/j.intimp.2024.113110] [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: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related mortality. Cancer poses a significant challenge to human health and remains a persistent and pressing issue. Schisandrin C is one of the active ingredients of Schisandra chinensis and has various biological and pharmacological activities. This study aimed to investigate the effects of Schisandrin C on lung cancer and the underlying mechanism involved. METHODS A network pharmacology strategy was used to screen the target genes and pathways involved in the relationship between Schisandrin and lung cancer. Next, a single-cell RNA sequencing (scRNA-seq) assay revealed the expression of genes specifically expressed in lung cancer epithelial cells. A549 cells were subsequently treated with Schisandrin C for 24 h or 48 h, cell viability was assessed via MTT and EdU staining experiments, and target gene expression was measured via RT-qPCR and immunofluorescence assays. Moreover, lung cancer patient tissues were observed via multiplex immunofluroscence staining. RESULTS AKT1, CA9, BRAF, EGFR, ERBB2 and PIK3CA were overlapping target genes for network pharmacology and the scRNA-seq strategy. In vitro, the RT-qPCR results indicated that Schisandrin C inhibited the mRNA expression of the AKT1, CA9, FASN, MMP1, EGFR and BRAF genes. In clinical samples from patients with lung cancer, the expression levels of CA9 and AKT1 were found to be significantly higher in lung tumor tissues than in the adjacent normal (TAN) tissues. Moreover, the administration of an AKT kinase inhibitor reversed the inhibitory effect of Schisandrin C on A549 cells proliferation, whereas the administration of a CA9 inhibitor failed to have a similar effect. CONCLUSIONS Schisandrin C effectively suppressed the proliferation and viability of A549 cells. Its mechanism was related to the inhibition of the AKT1 signaling pathway.
Collapse
Affiliation(s)
| | - Shengyang Xie
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Li Li
- Zhejiang Hospital, Hangzhou 310013, China
| | - Zhengcheng Liu
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Wencheng Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China.
| |
Collapse
|
7
|
Zhou R, Liu Z, Wu T, Pan X, Li T, Miao K, Li Y, Hu X, Wu H, Hemmings AM, Jiang B, Zhang Z, Liu N. Machine learning-aided discovery of T790M-mutant EGFR inhibitor CDDO-Me effectively suppresses non-small cell lung cancer growth. Cell Commun Signal 2024; 22:585. [PMID: 39639305 PMCID: PMC11619116 DOI: 10.1186/s12964-024-01954-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) T790M mutation often occurs during long durational erlotinib treatment of non-small cell lung cancer (NSCLC) patients, leading to drug resistance and disease progression. Identification of new selective EGFR-T790M inhibitors has proven challenging through traditional screening platforms. With great advances in computer algorithms, machine learning improved the screening rates of molecules at full chemical spaces, and these molecules will present higher biological activity and targeting efficiency. METHODS An integrated machine learning approach, integrated by Bayesian inference, was employed to screen a commercial dataset of 70,413 molecules, identifying candidates that selectively and efficiently bind with EGFR harboring T790M mutation. In vitro cellular assays and molecular dynamic simulations was used for validation. EGFR knockout cell line was generated for cross-validation. In vivo xenograft moues model was constructed to investigate the antitumor efficacy of CDDO-Me. RESULTS Our virtual screening and subsequent in vitro testing successfully identified CDDO-Me, an oleanolic acid derivative with anti-inflammatory activity, as a potent inhibitor of NSCLC cancer cells harboring the EGFR-T790M mutation. Cellular thermal shift assay and molecular dynamic simulation validated the selective binding of CDDO-Me to T790M-mutant EGFR. Further experimental results revealed that CDDO-Me induced cellular apoptosis and caused cell cycle arrest through inhibiting the PI3K-Akt-mTOR axis by directly targeting EGFR protein, cross-validated by sgEGFR silencing in H1975 cells. Additionally, CDDO-Me could dose-depended suppress the tumor growth in a H1975 xenograft mouse model. CONCLUSION CDDO-Me induced apoptosis and caused cell cycle arrest by inhibiting the PI3K-Akt-mTOR pathway, directly targeting the EGFR protein. In vivo studies in a H1975 xenograft mouse model demonstrated dose-dependent suppression of tumor growth. Our work highlights the application of machine learning-aided drug screening and provides a promising lead compound to conquer the drug resistance of NSCLC.
Collapse
Affiliation(s)
- Rui Zhou
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Ziqian Liu
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Tongtong Wu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475000, China
| | - Xianwei Pan
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Tongtong Li
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Kaiting Miao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475000, China
| | - Yuru Li
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaohui Hu
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Haigang Wu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475000, China
| | - Andrew M Hemmings
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Beier Jiang
- Naval Medicine Center of PLA, Naval Military University, Shanghai, 201306, China.
| | - Zhenzhen Zhang
- Naval Medicine Center of PLA, Naval Military University, Shanghai, 201306, China.
| | - Ning Liu
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China.
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, 201306, China.
| |
Collapse
|
8
|
Wei X, Zhang G, Liu Q, Niu Z, Chu C, Liu C, Wang K, Li L, Wang R, Cui W, Xu H, Liu C, Wang Y, An L. Almonertinib and alflutinib show novel inhibition on rare EGFR S768I mutant cells. Clin Transl Oncol 2024; 26:3100-3115. [PMID: 38814541 DOI: 10.1007/s12094-024-03494-5] [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: 02/01/2024] [Accepted: 04/01/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE EGFR classical mutations respond well to EGFR tyrosine kinase inhibitors. However, it is uncertain whether currently available EGFR-TKIs are effective against rare EGFR mutations and compound mutations. Herein, the effectiveness of almonertinib and alflutinib, the third-generation tyrosine kinase inhibitors developed in China, on rare EGFR S768I mutations and compound mutations is identified. METHODS In this study, using CRISPR method, four EGFR S768I mutation cell lines were constructed, and the sensitivity of EGFR to almonertinib and alflutinib was tested, with positive controls being the 1st (gefitinib), 2nd (afatinib), and 3rd (osimertinib) generation drugs. RESULTS The present results indicate that almonertinib and alflutinib can effectively inhibit cell viability and proliferation in rare EGFR S768I mutations through the ERK or AKT pathways in a time-dependent manner, by blocking the cell cycle and inhibiting apoptosis. CONCLUSIONS These findings suggest that almonertinib and alflutinib may be potential therapeutic options for non-small cell lung cancer patients with the EGFR S768I mutation.
Collapse
Affiliation(s)
- Xiangkai Wei
- Department of Anesthesiology, The First Affiliated Hospital of Henan University, Henan University, No. 357, Ximen Avenue, Kaifeng, 475000, Henan, China
| | - Guoliang Zhang
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
- Institutes of Traditional Chinese Medicine, School of Pharmacy, Henan University, Kaifeng, 475000, Henan, China
| | - Qian Liu
- Department of Anesthesiology, The First Affiliated Hospital of Henan University, Henan University, No. 357, Ximen Avenue, Kaifeng, 475000, Henan, China
| | - Zhiyuan Niu
- Department of Anesthesiology, The First Affiliated Hospital of Henan University, Henan University, No. 357, Ximen Avenue, Kaifeng, 475000, Henan, China
| | - Chunhong Chu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
- Institutes of Traditional Chinese Medicine, School of Pharmacy, Henan University, Kaifeng, 475000, Henan, China
| | - Chenxue Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Ke Wang
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Lanxin Li
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Rui Wang
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Wenrui Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Huixia Xu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Chenyang Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China
| | - Ying Wang
- Department of Anesthesiology, The First Affiliated Hospital of Henan University, Henan University, No. 357, Ximen Avenue, Kaifeng, 475000, Henan, China.
| | - Lei An
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, No. 115, Ximen Avenue, Kaifeng, 475000, China.
- Institutes of Traditional Chinese Medicine, School of Pharmacy, Henan University, Kaifeng, 475000, Henan, China.
| |
Collapse
|
9
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-x] [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: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
10
|
Wang X, Qin Z, Qiu W, Xu K, Bai Y, Zeng B, Ma Y, Yang S, Shi Y, Fan Y. Novel EGFR inhibitors against resistant L858R/T790M/C797S mutant for intervention of non-small cell lung cancer. Eur J Med Chem 2024; 277:116711. [PMID: 39094277 DOI: 10.1016/j.ejmech.2024.116711] [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: 06/17/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
To overcome C797S mutation, the latest and most common resistance mechanism in the clinical treatment of third-generation EGFR inhibitor, a novel series of substituted 6-(2-aminopyrimidine)-indole derivatives were designed and synthesized. Through the structure-activity relationship (SAR) study, compound 11eg was identified as a novel and potent EGFR L858R/T790M/C797S inhibitor (IC50 = 0.053 μM) but had a weak effect on EGFRWT (IC50 = 1.05 μM). 11eg significantly inhibited the proliferation of the non-small cell lung cancer (NSCLC) cells harboring EGFRL858R/T790M/C797S with an IC50 of 0.052 μM. 11eg also showed potent inhibitory activity against other NSCLC cell lines harboring main EGFR mutants. Furthermore, 11eg exhibited much superior activity in arresting cell cycle and inducing apoptosis of NSCLC cells with mutant EGFRC797S. It blocked cellular EGFR signaling. Importantly, 11eg markedly suppressed the tumor growth in in vivo xenograft mouse model with good safety. Additionally, 11eg displayed good microsomal stability. These results demonstrated the potential of 11eg with novel scaffold as a promising lead compound targeting EGFRC797S to guide in-depth structural optimization.
Collapse
Affiliation(s)
- Xiaoxue Wang
- Eye Institute, Nankai University, 94 Weijin Road, Tianjin, 300071, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Zhongxiang Qin
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Wenrui Qiu
- Tianjin Normal University, No.393, Extension of Bin Shui West Road, Xi Qing District, Tianjin, 300387, China
| | - Kejia Xu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yuting Bai
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Beilei Zeng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yakun Ma
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Shuang Yang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Yi Shi
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Yan Fan
- Eye Institute, Nankai University, 94 Weijin Road, Tianjin, 300071, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
11
|
Imam IA, Al Adawi S, Liu X, Ellingson S, Brainson CF, Moseley HNB, Zinner R, Zhang S, Shao Q. L858R/L718Q and L858R/L792H Mutations of EGFR Inducing Resistance Against Osimertinib by Forming Additional Hydrogen Bonds. Proteins 2024. [PMID: 39494831 DOI: 10.1002/prot.26761] [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: 07/16/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Acquired resistance to first-line treatments in various cancers both promotes cancer recurrence as well as limits effective treatment. This is true for epidermal growth factor receptor (EGFR) mutations, for which secondary EGFR mutations are one of the principal mechanisms conferring resistance to the covalent inhibitor osimertinib. Thus, it is very important to develop a deeper understanding of the secondary mutational resistance mechanisms associated with EGFR mutations arising in tumors treated with osimertinib to expedite the development of innovative therapeutic drugs to overcome acquired resistance. This work uses all-atom molecular dynamics (MD) simulations to investigate the conformational variation of two reported EGFR mutants (L858R/L718Q and L858R/L792H) that resist osimertinib. The wild-type EGFR kinase domain and the L858R mutant are used as the reference. Our MD simulation results revealed that both the L718Q and L792H secondary mutations induce additional hydrogen bonds between the residues in the active pocket and the residues with the water molecules. These additional hydrogen bonds reduce the exposure area of C797, the covalent binding target of osimertinib. The additional hydrogen bonds also influence the binding affinity of the EGFR kinase domain by altering the secondary structure and flexibility of the amino acid residues in the domain. Our work highlights how the two reported mutations may alter both residue-residue and residue-solvent hydrogen bonds, affecting protein binding properties, which could be helpful for future drug discovery.
Collapse
Affiliation(s)
- Ibrahim A Imam
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky, USA
| | - Shatha Al Adawi
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Sally Ellingson
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
- College of Medicine, Division of Biomedical Informatics University of Kentucky, Lexington, Kentucky, USA
| | - Christine F Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Hunter N B Moseley
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Ralph Zinner
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Shulin Zhang
- College of Medicine, Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Qing Shao
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
12
|
Xiao X, Xu R, Lu J, Xin B, Wang C, Zhu K, Zhang H, Chen X. The potential role of next-generation sequencing in identifying MET amplification and disclosing resistance mechanisms in NSCLC patients with osimertinib resistance. Front Oncol 2024; 14:1470827. [PMID: 39497720 PMCID: PMC11532092 DOI: 10.3389/fonc.2024.1470827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
Purposes Osimertinib, one of the third-generation EGFR-tyrosine kinase inhibitors (TKIs) designed to target EGFR T790M mutation, significantly improves the prognosis of lung cancer. However, drug resistance still happens and MET amplification is responsible for one of the main causes. Fluorescence in situ hybridization (FISH) is the gold standard for MET amplification detection, but fundamentally limited by observer subjectivity. Herein, we assessed the value of next-generation sequencing (NGS) method in MET amplification detection in non-small cell lung cancer (NSCLC), as well as revealed the mutation profiling of NSCLC patients with osimertinib resistance to provide some valuable clues to the mechanisms of resistance. Methods A total of 317 cancer tissue samples from 317 NSCLC patients at time of progression following osimertinib were submitted to NGS and only 96 tissues were tested by FISH simultaneously. With FISH results as gold standard, enumeration algorithm was applied to establish the optimal model for identifying MET amplification using gene copy number (GCN) data. Results The optimal model for identifying MET amplification was constructed based on the GCN of MET, BRAF, CDK6 and CYP3A4, which achieved a 74.0% overall agreement with FISH and performed well in identifying MET amplification except polysomy with a sensitivity of 85.7% and a specificity of 93.9%. The inconsistency between NGS and FISH occurred mainly in polysomy subtype, while MET GCN ≥ 5 could be reliably recognized by NGS. Moreover, the most frequently mutated genes in NSCLC patients with osimertinib resistance were EGFR (59.94%), followed by TP53 (43.85%), NRG1 (9.46%), PIK3CA (6.31%), and ATM (5.36%). The known resistance mechanisms, including MET amplification, EGFR (C797S, L718Q/R), TP53, CDK4, CDK6, CDKN2A, BRAF, KRAS, NRAS and PIK3CA mutations were also disclosed in our cohort. Conclusions NGS assay can achieve a high concordance with FISH in MET amplification detection and has advantages in portraying various genetic alterations, which is of worthy in clinical promotion.
Collapse
Affiliation(s)
- Xiao Xiao
- School of Physics, Changchun University of Science and Technology, Changchun, China
- Research & Development Department, Shanghai Rightongene Biotechnology Co., Ltd., Shanghai, China
| | - Ren Xu
- School of Physics, Changchun University of Science and Technology, Changchun, China
- Research & Development Department, Shanghai Rightongene Biotechnology Co., Ltd., Shanghai, China
| | - Jun Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beibei Xin
- Research & Development Department, Shanghai Rightongene Biotechnology Co., Ltd., Shanghai, China
| | - Chenyang Wang
- Research & Development Department, Shanghai Rightongene Biotechnology Co., Ltd., Shanghai, China
| | - Kexin Zhu
- Research & Development Department, Shanghai Rightongene Biotechnology Co., Ltd., Shanghai, China
| | - Hao Zhang
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Xinyu Chen
- School of Physics, Changchun University of Science and Technology, Changchun, China
| |
Collapse
|
13
|
Ou X, Gao G, Habaz IA, Wang Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm (Beijing) 2024; 5:e694. [PMID: 39184861 PMCID: PMC11344283 DOI: 10.1002/mco2.694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Tyrosine kinase inhibitor (TKI)-targeted therapy has revolutionized cancer treatment by selectively blocking specific signaling pathways crucial for tumor growth, offering improved outcomes with fewer side effects compared with conventional chemotherapy. However, despite their initial effectiveness, resistance to TKIs remains a significant challenge in clinical practice. Understanding the mechanisms underlying TKI resistance is paramount for improving patient outcomes and developing more effective treatment strategies. In this review, we explored various mechanisms contributing to TKI resistance, including on-target mechanisms and off-target mechanisms, as well as changes in the tumor histology and tumor microenvironment (intrinsic mechanisms). Additionally, we summarized current therapeutic approaches aiming at circumventing TKI resistance, including the development of next-generation TKIs and combination therapies. We also discussed emerging strategies such as the use of dual-targeted antibodies and PROteolysis Targeting Chimeras. Furthermore, we explored future directions in TKI-targeted therapy, including the methods for detecting and monitoring drug resistance during treatment, identification of novel targets, exploration of dual-acting kinase inhibitors, application of nanotechnologies in targeted therapy, and so on. Overall, this review provides a comprehensive overview of the challenges and opportunities in TKI-targeted therapy, aiming to advance our understanding of resistance mechanisms and guide the development of more effective therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Xuejin Ou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China HospitalSichuan UniversityChengduChina
| | - Inbar A. Habaz
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
14
|
Pan X, Zhou X. Long term survival achieved through combination of almonertinib and pyrotinib in EGFR-mutant/HER2-amplified advanced NSCLC patient: a case report and literature review. Front Oncol 2024; 14:1397238. [PMID: 39184039 PMCID: PMC11341367 DOI: 10.3389/fonc.2024.1397238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
Backgroud Human epithelial growth factor receptor 2 (HER2) amplification is an important mechanism of acquired resistance to anti-epidermal growth factor receptor (EGFR) therapy in non-small cell lung cancer (NSCLC) patients. For patients with both EGFR mutation and HER2 amplification, there is currently no unified standard treatment, and further exploration is needed on how to choose the therapy. Methods and results A female NSCLC patient developed bone and brain metastases 14 and 42 months after radical surgery, respectively. The second genetic sequencing detected EGFR L858R mutation and HER2 amplification, and therefore initiated treatment with almonertinib and pyrotinib. The patient achieved partial remission and did not show any further progression during the follow-up period. Conclusion For NSCLC patients with both EGFR mutation and HER2 amplification, the combination of almonertinib and pyrotinib is a valuable therapy that can continuously reduce tumor burden and achieve long-term survival.
Collapse
Affiliation(s)
| | - Xiao Zhou
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
15
|
Dai S, Liu Y, Liu Z, Li R, Luo F, Li Y, Dai L, Peng X. Cancer-associated fibroblasts mediate resistance to anti-EGFR therapies in cancer. Pharmacol Res 2024; 206:107304. [PMID: 39002870 DOI: 10.1016/j.phrs.2024.107304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Over the last decade, epidermal growth factor receptor (EGFR)-targeted therapies have transformed the treatment landscape for patients with advanced solid tumors. Despite these advances, resistance to anti-EGFR therapies is still a significant clinical challenge. While cell-autonomous mechanisms of resistance are well-documented, they do not fully elucidate the complexity of drug resistance. Cancer-associated fibroblasts (CAFs), key mediators within the tumor microenvironment (TME), have emerged as pivotal players in cancer progression and chemoresistance. Recent evidence implicates CAFs in resistance to anti-EGFR therapies, suggesting they may undermine treatment efficacy. This review synthesizes current data, highlighting the critical role of CAFs in resistance pathogenesis and summarizing recent therapeutic strategies targeting CAFs. We underscore the challenges and advocate for the exploration of CAFs as a potential dual-targeted approach.
Collapse
Affiliation(s)
- Shuang Dai
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingtong Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610041, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China
| | - Ruidan Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China.
| |
Collapse
|
16
|
Song Y, Dong QQ, Ni YK, Xu XL, Chen CX, Chen W. Nano-Proteolysis Targeting Chimeras (Nano-PROTACs) in Cancer Therapy. Int J Nanomedicine 2024; 19:5739-5761. [PMID: 38882545 PMCID: PMC11180470 DOI: 10.2147/ijn.s448684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules that have the capability to induce specific protein degradation. While playing a revolutionary role in effectively degrading the protein of interest (POI), PROTACs encounter certain limitations that impede their clinical translation. These limitations encompass off-target effects, inadequate cell membrane permeability, and the hook effect. The advent of nanotechnology presents a promising avenue to surmount the challenges associated with conventional PROTACs. The utilization of nano-proteolysis targeting chimeras (nano-PROTACs) holds the potential to enhance specific tissue accumulation, augment membrane permeability, and enable controlled release. Consequently, this approach has the capacity to significantly enhance the controllable degradation of target proteins. Additionally, they enable a synergistic effect by combining with other therapeutic strategies. This review comprehensively summarizes the structural basis, advantages, and limitations of PROTACs. Furthermore, it highlights the latest advancements in nanosystems engineered for delivering PROTACs, as well as the development of nano-sized PROTACs employing nanocarriers as linkers. Moreover, it delves into the underlying principles of nanotechnology tailored specifically for PROTACs, alongside the current prospects of clinical research. In conclusion, the integration of nanotechnology into PROTACs harbors vast potential in enhancing the anti-tumor treatment response and expediting clinical translation.
Collapse
Affiliation(s)
- Yue Song
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, People’s Republic of China
| | - Qing-Qing Dong
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Yi-Ke Ni
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Chao-Xiang Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Wei Chen
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
17
|
Du Y, Shi S, Shu C, He Y, Xu W, Wu D, Tian Y, Kong M, He J, Xie W, Qiu Y, Xu Y, Zou Y, Zhu Q. Discovery of novel EGFR-PROTACs capable of degradation of multiple EGFR-mutated proteins. Eur J Med Chem 2024; 272:116489. [PMID: 38759458 DOI: 10.1016/j.ejmech.2024.116489] [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: 03/19/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
Although three generations of Epidermal growth factor receptor (EGFR) - TK inhibitors have been approved for the treatment of Non-small-cell lung cancers (NSCLC), their clinical application is still largely hindered by acquired drug resistance mediated new EGFR mutations and side effects. The Proteolysis targeting chimera (PROTAC) technology has the potential to overcome acquired resistance from mutant EGFR through a novel mechanism of action. In this study, we developed the candidate degrader IV-3 by structural modifications of the lead compound 13, which exhibited limited antiproliferative activity against HCC-827 cells. Compared to compound 13, IV-3 exhibited remarkable anti-proliferative activity against HCC-827 cells, NCI-H1975 cells, and NCI-H1975-TM cells (IC50 = 0.009 μM, 0.49 μM and 3.24 μM, respectively), as well as significantly inducing degradation of EGFR protein in these cell lines (DC50 = 17.93 nM, 0.25 μM and 0.63 μM, respectively). Further investigations confirmed that IV-3 exhibited superior anti-tumor activity in all xenograft tumor models through the degradation of mutant EGFR protein. Moreover, IV-3 showed no inhibitory activity against A431 and A549 cells expressing wild-type EGFR, thereby eliminating potential toxic side effects emerging from wild-type EGFR inhibition. Overall, our study provides promising insights into EGFR-PROTACs as a potential therapeutic strategy against EGFR-acquired mutation.
Collapse
Affiliation(s)
- Yu Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Shi Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China; Jiangsu Lianhuan Pharmaceutical Co., Ltd, Yangzhou 225000, China
| | - Chen Shu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yezi He
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wangyang Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Daochen Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yushu Tian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Mingyang Kong
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jiahuan He
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wenhui Xie
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yijia Qiu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yungen Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Yi Zou
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Qihua Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
18
|
Xu J, Tian L, Qi W, Lv Q, Wang T. Advancements in NSCLC: From Pathophysiological Insights to Targeted Treatments. Am J Clin Oncol 2024; 47:291-303. [PMID: 38375734 PMCID: PMC11107893 DOI: 10.1097/coc.0000000000001088] [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] [Indexed: 02/21/2024]
Abstract
With the global incidence of non-small cell lung cancer (NSCLC) on the rise, the development of innovative treatment strategies is increasingly vital. This review underscores the pivotal role of precision medicine in transforming NSCLC management, particularly through the integration of genomic and epigenomic insights to enhance treatment outcomes for patients. We focus on the identification of key gene mutations and examine the evolution and impact of targeted therapies. These therapies have shown encouraging results in improving survival rates and quality of life. Despite numerous gene mutations being identified in association with NSCLC, targeted treatments are available for only a select few. This paper offers an exhaustive analysis of the pathogenesis of NSCLC and reviews the latest advancements in targeted therapeutic approaches. It emphasizes the ongoing necessity for research and development in this domain. In addition, we discuss the current challenges faced in the clinical application of these therapies and the potential directions for future research, including the identification of novel targets and the development of new treatment modalities.
Collapse
Affiliation(s)
- Jianan Xu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine
| | - Lin Tian
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| | - Wenlong Qi
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| | - Qingguo Lv
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| | - Tan Wang
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| |
Collapse
|
19
|
Li Z, Zou W, Yuan J, Zhong Y, Fu Z. Gender differences in adverse events related to Osimertinib: a real-world pharmacovigilance analysis of FDA adverse event reporting system. Expert Opin Drug Saf 2024; 23:763-770. [PMID: 37515501 DOI: 10.1080/14740338.2023.2243220] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE We analyze and identify the signals of gender differences in adverse events (ADEs) related to Osimertinib and provide reference for clinical implementation of individualized drug use. METHODS ADE reports of Osimertinib received from FAERS database from the first quarter of 2016 to the fourth quarter of 2022 were extracted. Reporting odds ratio (ROR) data analysis strategy was used for mining of signal strength that represents gender differences in ADEs related to Osimertinib. RESULTS The number of Osimertinib ADE reports included in the analysis was 7968 in females and 7570 in males, respectively. According to ROR, men were more likely to develop pneumonia aspiration, lung infection, interstitial lung disease, pulmonary toxicity, dyspnea, ventricular extrasystoles, and pulmonary thrombosis, while women were more likely to develop cardiac failure congestive, stomatitis, diarrhea, muscle spasms, nail disorder, onycholysis, skin disorder, dry skin, and rash. CONCLUSION Gender differences existed in ADE signals related to Osimertinib. The higher risk of ADEs in male patients was lung diseases that seem more serious than those nail toxicities or skin problems that occurred in female patients. In order to ensure the safety of medication, we should be alert to the differences between different genders and take corresponding preventive measures to reduce the occurrence of serious ADEs.
Collapse
Affiliation(s)
- Zhiping Li
- Department of Pharmacy, Dongguan Binhaiwan Center Hospital, Dongguan, China
| | - Wenbin Zou
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Yuan
- Department of Pharmacy, Dongguan Binhaiwan Center Hospital, Dongguan, China
| | - Yunxiang Zhong
- Department of Pharmacy, Dongguan Binhaiwan Center Hospital, Dongguan, China
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Latham BD, Geffert RM, Jackson KD. Kinase Inhibitors FDA Approved 2018-2023: Drug Targets, Metabolic Pathways, and Drug-Induced Toxicities. Drug Metab Dispos 2024; 52:479-492. [PMID: 38286637 PMCID: PMC11114602 DOI: 10.1124/dmd.123.001430] [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: 10/05/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024] Open
Abstract
Small molecule kinase inhibitors are one of the fastest growing classes of drugs, which are approved by the US Food and Drug Administration (FDA) for cancer and noncancer indications. As of September 2023, there were over 70 FDA-approved small molecule kinase inhibitors on the market, 42 of which were approved in the past five years (2018-2023). This minireview discusses recent advances in our understanding of the pharmacology, metabolism, and toxicity profiles of recently approved kinase inhibitors with a central focus on tyrosine kinase inhibitors (TKIs). In this minireview we discuss the most common therapeutic indications and molecular target(s) of kinase inhibitors FDA approved 2018-2023. We also describe unique aspects of the metabolism, bioactivation, and drug-drug interaction (DDI) potential of kinase inhibitors; discuss drug toxicity concerns related to kinase inhibitors, such as drug-induced liver injury; and highlight clinical outcomes and challenges relevant to TKI therapy. Case examples are provided for common TKI targets, metabolism pathways, DDI potential, and risks for serious adverse drug reactions. The minireview concludes with a discussion of perspectives on future research to optimize TKI therapy to maximize efficacy and minimize drug toxicity. SIGNIFICANCE STATEMENT: This minireview highlights important aspects of the clinical pharmacology and toxicology of small molecule kinase inhibitors FDA approved 2018-2023. We describe key advances in the therapeutic indications and molecular targets of TKIs. The major metabolism pathways and toxicity profiles of recently approved TKIs are discussed. Clinically relevant case examples are provided that demonstrate the risk for hepatotoxic drug interactions involving TKIs and coadministered drugs.
Collapse
Affiliation(s)
- Bethany D Latham
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Raeanne M Geffert
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
21
|
Xu J, Yang Y, Gao Z, Song T, Ma Y, Yu X, Shi C. Distinguishing EGFR mutation molecular subtypes based on MRI radiomics features of lung adenocarcinoma brain metastases. Clin Neurol Neurosurg 2024; 240:108258. [PMID: 38552362 DOI: 10.1016/j.clineuro.2024.108258] [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: 02/18/2024] [Accepted: 03/23/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVE To explore the feasibility of identifying epidermal growth factor receptor (EGFR) mutation molecular subtypes in primary lesions based on the radiomics features of lung adenocarcinoma brain metastases using magnetic resonance imaging (MRI). METHODS We retrospectively analyzed clinical, imaging, and genetic testing data of patients with lung adenocarcinoma with EGFR gene mutations who had brain metastases. Three-dimensional radiomics features were extracted from contrast-enhanced T1-weighted images. The volume of interest was delineated and normalized using Z-score, dimensionality reduction was performed using principal component analysis, feature selection using Relief, and radiomics model construction using adaptive boosting as a classifier. Data were randomly divided into training and testing datasets at an 8:2 ratio. Five-fold cross-validation was conducted in the training set to select the optimal radiomics features and establish a predictive model for distinguishing between exon 19 deletion (19Del) and exon 21 L858R point mutation (21L858R), the two most common EGFR gene mutations. The testing set was used for external validation of the models. Model performance was evaluated using receiver operating characteristic curve and decision curve analyses. RESULTS Overall, 86 patients with 228 brain metastases were included. Patient age was identified as an independent predictor for distinguishing between 19Del and 21L858R. The area under the curve (AUC) values of the radiomics model in the training and testing datasets were 0.895 (95% confidence interval [CI]: 0.850-0.939) and 0.759 (95% CI: 0.0.614-0.903), respectively. The AUC for diagnosis of all cases using a combined model of age and radiomics was 0.888 (95% CI: 0.846-0.930), slightly higher than that of the radiomics model alone (0.866, 95% CI: 0.820-0.913), but without statistical significance (p=0.1626). In the decision curve analysis, both models demonstrated clinical net benefits. CONCLUSIONS The radiomics model based on MRI of lung adenocarcinoma brain metastases could distinguish between EGFR 19Del and 21L858R mutations in the primary lesion.
Collapse
Affiliation(s)
- Jiali Xu
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China; Department of Medical Imaging Diagnosis, School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China.
| | - Yuqiong Yang
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China; School of Graduate, Bengbu Medical University, Bengbu, Anhui 233030,China
| | - Zhizhen Gao
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Tao Song
- Vascular Surgery Department, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Yichuan Ma
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Xiaojun Yu
- Department of Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Changzheng Shi
- Department of Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| |
Collapse
|
22
|
Ferro A, Marinato GM, Mulargiu C, Marino M, Pasello G, Guarneri V, Bonanno L. The study of primary and acquired resistance to first-line osimertinib to improve the outcome of EGFR-mutated advanced Non-small cell lung cancer patients: the challenge is open for new therapeutic strategies. Crit Rev Oncol Hematol 2024; 196:104295. [PMID: 38382773 DOI: 10.1016/j.critrevonc.2024.104295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
The development of targeted therapy in epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC) patients has radically changed their clinical perspectives. Current first-line standard treatment for advanced disease is commonly considered third-generation tyrosine kinase inhibitors (TKI), osimertinib. The study of primary and acquired resistance to front-line osimertinib is one of the main burning issues to further improve patients' outcome. Great heterogeneity has been depicted in terms of duration of clinical benefit and pattern of progression and this might be related to molecular factors including subtypes of EGFR mutations and concomitant genetic alterations. Acquired resistance can be categorized into two main classes: EGFR-dependent and EGFR-independent mechanisms and specific pattern of progression to first-line osimertinib have been demonstrated. The purpose of the manuscript is to provide a comprehensive overview of literature about molecular resistance mechanisms to first-line osimertinib, from a clinical perspective and therefore in relationship to emerging therapeutic approaches.
Collapse
Affiliation(s)
- Alessandra Ferro
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Gian Marco Marinato
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Cristiana Mulargiu
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Monica Marino
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Giulia Pasello
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Valentina Guarneri
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Laura Bonanno
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy.
| |
Collapse
|
23
|
Bai H, Zhou Y, Liu W, Xu WY, Cheng L, Huo Y, Ji H, Xiong L. Genetic mutation profiling reveals biomarkers for targeted therapy efficacy and prognosis in non-small cell lung cancer. Heliyon 2024; 10:e27633. [PMID: 38496877 PMCID: PMC10944256 DOI: 10.1016/j.heliyon.2024.e27633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction The genetic heterogeneity of non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations may affect clinical responses and outcomes to EGFR tyrosine kinase inhibitors (EGFR-TKIs). This study aims to investigate the genomic factors that influence the efficacy and clinical outcomes of first-line, second-line and third-line treatments in NSCLC and explore the heterogeneity of resistance mechanisms. Materials and methods This real-world study comprised 65 patients with EGFR mutant NSCLC. Molecular alterations were detected using a customized DNA panel before and after administering targeted therapy. The efficacy and prognosis of each treatment line were evaluated. Results In first-generation EGFR-TKIs treatment, gefitinib showed favorable efficacy compared to icotinib and erlotinib, particularly in patients with EGFR L858R mutations. The resistance mechanisms to first-generation EGFR-TKIs varied among different EGFR mutation cohorts and different first-generation EGFR-TKIs. In second-line EGFR-TKIs treatment, EPH receptor A3 (EPHA3), IKAROS family zinc finger 1 (IKZF1), p21 (RAC1) activated kinase 5 (PAK5), DNA polymerase epsilon, catalytic subunit (POLE), RAD21 cohesin complex component (RAD21) and RNA binding motif protein 10 (RBM10) mutations were markedly associated with poorer progression-free survival (PFS). Notably, EPHA3, IKZF1 and RBM10 were identified as independent predictors of PFS. The mechanisms of osimertinib resistance exhibited heterogeneity, with a higher proportion of non-EGFR-dependent resistant mutations. In third-line treatments, the combination of osimertinib and anlotinib demonstrated superior efficacy compared to other regimens. Glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A) mutation was an independent risk indicator of shorter OS following third-line treatments. Conclusions Comprehending the tumor evolution in NSCLC is advantageous for assessing the efficacy and prognosis at each stage of treatment, providing valuable insights to guide personalized treatment decisions for patients.
Collapse
Affiliation(s)
- Hao Bai
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yan Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Wanting Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | | | - Lei Cheng
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | | | - Hao Ji
- Department of Respiratory and Critical Care Medicine, Department of Healthcare Associated Infection Management, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Liwen Xiong
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| |
Collapse
|
24
|
Wang YT, Yang PC, Zhang JY, Sun JF. Synthetic Routes and Clinical Application of Representative Small-Molecule EGFR Inhibitors for Cancer Therapy. Molecules 2024; 29:1448. [PMID: 38611728 PMCID: PMC11012680 DOI: 10.3390/molecules29071448] [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: 12/07/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The epidermal growth factor receptor (EGFR) plays a pivotal role in cancer therapeutics, with small-molecule EGFR inhibitors emerging as significant agents in combating this disease. This review explores the synthesis and clinical utilization of EGFR inhibitors, starting with the indispensable role of EGFR in oncogenesis and emphasizing the intricate molecular aspects of the EGFR-signaling pathway. It subsequently provides information on the structural characteristics of representative small-molecule EGFR inhibitors in the clinic. The synthetic methods and associated challenges pertaining to these compounds are thoroughly examined, along with innovative strategies to overcome these obstacles. Furthermore, the review discusses the clinical applications of FDA-approved EGFR inhibitors such as erlotinib, gefitinib, afatinib, and osimertinib across various cancer types and their corresponding clinical outcomes. Additionally, it addresses the emergence of resistance mechanisms and potential counterstrategies. Taken together, this review aims to provide valuable insights for researchers, clinicians, and pharmaceutical scientists interested in comprehending the current landscape of small-molecule EGFR inhibitors.
Collapse
Affiliation(s)
- Ya-Tao Wang
- First People’s Hospital of Shangqiu, Shangqiu 476100, China
| | - Peng-Cheng Yang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China;
| | - Jing-Yi Zhang
- College of Chemistry and Chemical Engineering, Zhengzhou Normal University, Zhengzhou 450044, China;
| | - Jin-Feng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China;
| |
Collapse
|
25
|
Zhen S, Jia Y, Zhao Y, Wang J, Zheng B, Liu T, Duan Y, Lv W, Wang J, Xu F, Liu Y, Zhang Y, Liu L. NEAT1_1 confers gefitinib resistance in lung adenocarcinoma through promoting AKR1C1-mediated ferroptosis defence. Cell Death Discov 2024; 10:131. [PMID: 38472205 DOI: 10.1038/s41420-024-01892-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Gefitinib is one of the most extensively utilized epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) for treating advanced lung adenocarcinoma (LUAD) patients harboring EGFR mutation. However, the emergence of drug resistance significantly compromised the clinical efficacy of EGFR-TKIs. Gaining further insights into the molecular mechanisms underlying gefitinib resistance holds promise for developing novel strategies to overcome the resistance and improve the prognosis in LUAD patients. Here, we identified that the inhibitory efficacy of gefitinib on EGFR-mutated LUAD cells was partially dependent on the induction of ferroptosis, and ferroptosis protection resulted in gefitinib resistance. Among the ferroptosis suppressors, aldo-keto reductase family 1 member C1 (AKR1C1) exhibited significant upregulation in gefitinib-resistant strains of LUAD cells and predicted poor progression-free survival (PFS) and overall survival (OS) of LUAD patients who received first-generation EGFR-TKI treatment. Knockdown of AKR1C1 partially reversed drug resistance by re-sensitizing the LUAD cells to gefitinib-mediated ferroptosis. The decreased expression of miR-338-3p contributed to the aberrant upregulation of AKR1C1 in gefitinib-resistant LUAD cells. Furthermore, upregulated long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1_1 (NEAT1_1) sponged miR-338-3p to neutralize its suppression on AKR1C1. Dual-luciferase reporter assay and miRNA rescue experiment confirmed the NEAT1_1/miR-338-3p/AKR1C1 axis in EGFR-mutated LUAD cells. Gain- and loss-of-function assays demonstrated that the NEAT1_1/miR-338-3p/AKR1C1 axis promoted gefitinib resistance, proliferation, migration, and invasion in LUAD cells. This study reveals the effects of NEAT1_1/miR-338-3p/AKR1C1 axis-mediated ferroptosis defence in gefitinib resistance in LUAD. Thus, targeting NEAT1_1/miR-338-3p/AKR1C1 axis might be a novel strategy for overcoming gefitinib resistance in LUAD harboring EGFR mutation.
Collapse
Affiliation(s)
- Shuman Zhen
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
- Department of Radiotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050017, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
| | - Yan Zhao
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
| | - Boyang Zheng
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Tianxu Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
| | - Wei Lv
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiaqi Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Fan Xu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, 067000, China
| | - Yueping Liu
- Department of Pathology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050017, China
| | - Yi Zhang
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China.
- China International Cooperation Laboratory of Stem Cell Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, 050017, China.
- Cancer Research Institute of Hebei Province, Shijiazhuang, 050017, China.
| |
Collapse
|
26
|
Lu G, Zhong H, Gao J, Zhang Y. Alginate microspheres encapsulating hox transcript antisense RNA siRNA regulate the Hedgehog-Gli1 pathway to alleviate epidermal growth factor receptor tyrosine kinase inhibitors resistance. J Biomater Appl 2024; 38:877-889. [PMID: 38261797 DOI: 10.1177/08853282241228667] [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] [Indexed: 01/25/2024]
Abstract
The long non-coding RNA HOTAIR and the Hedgehog-Gli1 signaling pathway are closely associated with tumor occurrence and drug resistance in various cancers. However, their specific roles in the development of EGFR-TKIs resistance in non-small cell carcinoma remain unclear. To address the issue of EGFR-TKIs resistance, this study utilized the electrospray method to prepare sodium alginate microspheres encapsulating HOTAIR siRNA (SA/HOTAIR siRNA) and investigated its effects on RNA interference (RNAi) in the gefitinib-resistant cell line PC9/GR. Furthermore, the study explored whether HOTAIR could modulate EGFR-TKIs resistance through the Hedgehog-GLi1 signaling pathway. The experimental results showed that sodium alginate (SA) microspheres demonstrated excellent biocompatibility with high encapsulation efficiency and drug-loading capacity, effectively enhancing the silencing efficiency of siRNA. HOTAIR siRNA significantly inhibited the proliferation, migration, and invasion abilities of PC9/GR cells while promoting apoptosis. Additionally, HOTAIR siRNA effectively suppressed tumor growth and downregulated the Hedgehog-GLi1 pathway and anti-apoptotic proteins, which were confirmed in animal experiments. Moreover, SA/HOTAIR siRNA exhibited superior inhibition of cellular and tumor functions compared to using HOTAIR siRNA alone. Clinical research findings indicated that monitoring the expression level of HOTAIR in the serum and urine samples of NSCLC patients before and after receiving EGFR-TKIs treatment can predict the efficacy of EGFR-TKIs to a certain extent. This study provided evidence that HOTAIR siRNA effectively mitigated the development of acquired resistance to EGFR-TKIs by inhibiting the Hedgehog-GLi1 pathway. Furthermore, it introduced a reliable and long-lasting drug delivery system for combating acquired resistance to EGFR-TKIs.
Collapse
Affiliation(s)
- Guojie Lu
- Department of Cardiothoracic Surgery, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiling Zhong
- Department of Cardiothoracic Surgery, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianwei Gao
- Department of Cardiothoracic Surgery, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaosen Zhang
- Department of Cardiothoracic Surgery, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
27
|
Jiang K, Wu L, Zheng X, Xu Y, Miao Q, Zheng X, Zhang L, Huang C, Lin G. Chemotherapy versus personalized therapy for EGFR mutant lung adenocarcinoma resistance to EGFR-tyrosine kinase inhibitors: a retrospective dual-center study. BMC Pulm Med 2024; 24:96. [PMID: 38402169 PMCID: PMC10893690 DOI: 10.1186/s12890-024-02905-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Advanced lung adenocarcinoma patients often develop resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs), leaving uncertainties regarding subsequent treatment strategies. Although personalized therapy targeting individual acquired resistances (ARs) shows promise, its efficacy has not been systematically compared with platinum-containing doublet chemotherapy, a widely accepted treatment after EGFR-TKIs failure. METHODS A retrospective dual-center study was conducted involving patients with advanced lung adenocarcinoma and EGFR mutations who developed resistance to EGFR-TKIs between January 2017 and December 2022. Eligible patients were adults aged 18 years or older with an Eastern Cooperative Oncology Group score of 0-1, normal organ function, and no prior chemotherapy. Patients were divided into the chemotherapy group (CG) or personalized therapy group (PG) based on the treatment received after disease progression. The primary endpoints were progression-free survival (PFS) and objective response rate (ORR). RESULTS Of the 144 patients enrolled, there were 53 patients in the PG and 91 patients in the CG. The PG acquired resistance to EGFR-TKIs through the MET amplification (27, 50%) and small cell lung cancer transformation (16, 30%) and 18% of them reported multiple resistance mechanisms. The ORR of the PG was similar to that of the CG (34% vs. 33%, P = 1.0) and the PFS of the PG patients was not statistically different from that of their CG counterparts [4.2 months (95% CI: 3.6-4.8 months) vs. 5.3 months (95% CI: 4.6-6.0 months), P = 0.77]. CONCLUSIONS These findings suggest that the therapeutic efficacy of chemotherapy approximates to that of personalized therapy, which signifies that chemotherapy is still a reliable choice for patients who develop resistance to EGFR-TKIs and that further research is awaited to explore the benefit of personalized treatment.
Collapse
Affiliation(s)
- Kan Jiang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lin Wu
- The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Xinlong Zheng
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yiquan Xu
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qian Miao
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xiaobin Zheng
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Longfeng Zhang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Cheng Huang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Gen Lin
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China.
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, China.
| |
Collapse
|
28
|
Hu L, Shi S, Song X, Ma F, Ji O, Qi B. Identification of novel aminopyrimidine derivatives for the treatment of mutant NSCLC. Eur J Med Chem 2024; 265:116074. [PMID: 38142512 DOI: 10.1016/j.ejmech.2023.116074] [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: 10/05/2023] [Revised: 12/10/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Starting from the binding mode of allosteric EGFR inhibitor JBJ-04-125-02 and the key pharmacophore of the third-generation EGFR inhibitors, we designed and synthesized a novel series of EGFR inhibitors, represented by (R)-N-(4-((2-aminopyrimidin-4-yl)amino)phenyl)-2-(5-(4-(4-methylpiperazin-1-yl)phenyl)-1-oxoisoindolin-2-yl)-2-phenylacetamide (6q). Docking study demonstrated that top compound 6q spanned orthosteric and allosteric sites of EGFR, and formed three key H-bonds with the residues Asp855, Lys745, and Met793 located in two sites. Biological evaluation indicated that compound 6q showed potential inhibitory activity against Ba/F3-EGFRL858R/T790M/C797S and Ba/F3-EGFRDel19/T790M/C797S cells, with IC50 values of 0.42 μM and 0.41 μM, respectively. Furthermore, compound 6q showed excellent activity against mutant NSCLC cell line NCI-H1975-EGFRL858R/T790M/C797S cells, with IC50 value of 0.82 μM which was superior to that of osimertinib (IC50 = 2.94 μM), JBJ-04-125-02 (IC50 = 3.66 μM), and coadministration of JBJ-04-125-02 and osimertinib (IC50 = 1.25 μM). Cell cycle arrest and cell apoptosis assay indicated that compound 6q could promote apoptosis of NCI-H1975-EGFRL858R/T790M/C797S cells at the concentration of 0.8 μM and no obvious cell cycle arrest was found.
Collapse
Affiliation(s)
- Liping Hu
- School of Bioengineering, Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis&Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Shengmin Shi
- School of Bioengineering, Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis&Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Xiaomeng Song
- School of Bioengineering, Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis&Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Fangli Ma
- School of Bioengineering, Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis&Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Oulian Ji
- School of Bioengineering, Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis&Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Baohui Qi
- School of Bioengineering, Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis&Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
29
|
Lu X, Yu R, Li Z, Yang M, Dai J, Liu M. JC-010a, a novel selective SHP2 allosteric inhibitor, overcomes RTK/non-RTK-mediated drug resistance in multiple oncogene-addicted cancers. Cancer Lett 2024; 582:216517. [PMID: 38101609 DOI: 10.1016/j.canlet.2023.216517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
Src homology 2 domain-containing phosphatase (SHP2) is a non-receptor protein phosphatase that transduces signals from upstream receptor tyrosine kinases (RTKs)/non-RTKs to Ras/MAPK pathway. Accumulating studies indicated that SHP2 is a critical mediator of resistance to current targeted therapies in multiple cancers. Here, we reported a novel SHP2 allosteric inhibitor JC-010a, which was highly selective to SHP2 and bound at the "tunnel" allosteric site of SHP2. The effect of JC-010a on combating RTK/non-RTK or MAPK inhibitors-induced acquired resistance was explored. Our study demonstrated that JC-010a monotherapy significantly inhibited the proliferation of cancer cells with different oncogenic drivers via inhibiting signaling through SHP2. Importantly, JC-010a abolished acquired resistance induced by targeted therapies: in KRAS-mutant cancers, JC-010a abrogated selumetinib-induced adaptive resistance mediated by RTK/SHP2; in BCR-ABL-driven leukemia cells, we demonstrated JC-010a inhibited BCR-ABL T315I mutation-mediated imatinib resistance and proposed a novel mechanism of JC-010a involving the disrupted co-interaction of SHP2, BCR-ABL, and Hsp90; in non-small cell lung cancer (NSCLC) cells, JC-010a inhibited both EGFR T790M/C797S mutation and alternate RTK-driven resistance to gefitinib or osimertinib; importantly, we first proposed a novel potential therapeutic strategy for RET-rearranged cancer, we confirmed that JC-010a monotherapy inhibited cell resistance to BLU-667, and JC-010a/BLU-667 combination prolonged anticancer response both in vivo and in vitro cancer models by inhibiting the alternate MET activation-induced RAS/MAPK reactivation, thereby promoting cancer cell apoptosis. These findings suggested that JC-010a was a novel selective SHP2 allosteric inhibitor, and combing JC-010a with current targeted therapy agents provided a promising therapeutic approach for clinical resistant cancers.
Collapse
Affiliation(s)
- Xuxiu Lu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Zhen Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Mengke Yang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Jiajia Dai
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
30
|
Zheng J, Dou Y, Huang D, Wang Y, Han R, Hu C, Zhu M, Lu C, Lin C, Wu D, Liu Y, Tang H, He T, Jiang W, He Y. Overall signature of acquired KRAS gene changes in advanced non-small cell lung cancer patient with EGFR-TKI resistance. Jpn J Clin Oncol 2024; 54:89-96. [PMID: 37721193 DOI: 10.1093/jjco/hyad123] [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: 05/04/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
OBJECTIVE Numerous scattered case studies continue to demonstrate a strong correlation between acquired KRAS mutations and epidermal growth factor receptor-tyrosine kinase inhibitor resistance in non-small cell lung cancer. However, the comprehensive understanding of the KRAS pathway following the failure of epidermal growth factor receptor-tyrosine kinase inhibitor therapy remains limited. METHODS We conducted a retrospective evaluation of the next generation sequencing data from 323 patients with advanced non-small cell lung cancer and EGFR-activating mutations after experiencing progression with epidermal growth factor receptor-tyrosine kinase inhibitor therapy. Our analysis specifically focused on the acquired changes to the KRAS gene. RESULTS Among the 323 patients with advanced non-small cell lung cancer and EGFR-activating mutations who experienced resistance to epidermal growth factor receptor-tyrosine kinase inhibitor therapy, 14 individuals (4.3%) developed resistance due to acquired KRAS alterations. Of these 14 patients, 10 cases (71.4%) were due to KRAS missense mutations, 1 case (7.2%) was due to KRAS gene fusion and 3 cases (21.4%) were due to KRAS amplification. Notably, we identified one newly demonstrated KRAS gene fusion (KRAS and LMNTD1), one KRAS G13D and one KRAS K117N. The emergence of acquired KRAS alterations was often accompanied by novel mutations and high tumor mutation burden, with TP53, CNKN2A, PIK3CA, MYC, STK11, CDK4, BRCA2 and ERBB2 being the most frequently observed concurrent mutations. The median progression-free survival and overall survival for the 14 patients were 5.2 and 7.3 months, respectively. Acquired KRAS missense variants were associated with significantly worse progression-free survival compared with other KRAS variant subtypes (P < 0.028). CONCLUSIONS This study provides significant evidence of the role of acquired KRAS variants in the development of resistance to epidermal growth factor receptor-tyrosine kinase inhibitor therapy. Our results contribute to the growing body of knowledge on the mutational profiles associated with resistance to epidermal growth factor receptor-tyrosine kinase inhibitor treatment. Furthermore, our study highlights the KRAS gene change as a significant mechanism of resistance to epidermal growth factor receptor-tyrosine kinase inhibitor therapy.
Collapse
Affiliation(s)
- Jie Zheng
- School of Medicine, Chongqing University, Chongqing, China
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Daijuan Huang
- School of Medicine, Chongqing University, Chongqing, China
| | - Yubo Wang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Mengxiao Zhu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Di Wu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Huan Tang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Tingting He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Weilin Jiang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yong He
- School of Medicine, Chongqing University, Chongqing, China
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
31
|
Nurisyah, Ramadhan DSF, Dewi R, asikin A, Daswi DR, adam A, Chaerunnimah, Sunarto, Rafika, Artati, Fakih TM. Targeting EGFR allosteric site with marine-natural products of Clathria Sp.: A computational approach. Curr Res Struct Biol 2024; 7:100125. [PMID: 38282755 PMCID: PMC10821630 DOI: 10.1016/j.crstbi.2024.100125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/30/2024] Open
Abstract
The EGFR-C797S resistance mutation to third-generation drugs has been overcome by fourth-generation inhibitors, allosteric inhibitors, namely EAI045 and has reached phase 3 clinical trials, so the Allosteric Site is currently an attractive target for development. In this study, researchers are interested in knowing the activity of metabolite compounds from marine natural ingredients Clathria Sp. against the Allosteric Site of EGFR computationally. The methods used include molecular docking using Autodock4 software and Molecular Dynamics simulation performed using GROMACS software. The research began with the preparation of metabolite samples from Clathria Sp. through the KnapSack database site and the preparation of EGFR receptors that have been complexed with allosteric inhibitors, namely proteins with PDB code 5D41. Each compound was docked to the Allosteric Site of the natural ligand and then molecular dynamics simulations were performed on the compound with the best docking energy compared to the natural ligand. From the docking results, the Clathrin_A compound showed the lowest binding energy compared to other metabolites, and the value was close to the natural ligand. Then from the molecular dynamics results, the clathrin_A compound shows good stability and resembles the natural ligand, which is analyzed through RMSD, RMSF, SASA, Rg, and PCA, and shows the binding free energy from MMPBSA analysis which is close to the natural ligand. It can be concluded, Clathrin_A compound has potential as an allosteric inhibitor.
Collapse
Affiliation(s)
- Nurisyah
- Department of Pharmacy, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | | | - Ratnasari Dewi
- Department of Pharmacy, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | - Asyhari asikin
- Department of Pharmacy, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | | | - Adriyani adam
- Department of Nutrition and Dietetics, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | - Chaerunnimah
- Department of Nutrition and Dietetics, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | - Sunarto
- Department of Nutrition and Dietetics, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | - Rafika
- Department of Medical Laboratory and Technology, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | - Artati
- Department of Medical Laboratory and Technology, Poltekkes Kemenkes Makassar, South Sulawesi, Indonesia
| | | |
Collapse
|
32
|
Metwally K, Abo-Dya NE. Pyrrolo[2,3-D]Pyrimidines as EGFR and VEGFR Kinase Inhibitors: A Comprehensive SAR Review. Curr Med Chem 2024; 31:5918-5936. [PMID: 37581522 DOI: 10.2174/0929867331666230815115111] [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: 05/12/2023] [Revised: 06/17/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023]
Abstract
Tyrosine kinases are implicated in a wide array of cellular physiological processes, including cell signaling. The discovery of the BCR-ABL tyrosine kinase inhibitor imatinib and its FDA approval in 2001 paved the way for the development of small molecule chemical entities of diverse structural backgrounds as tyrosine kinase inhibitors for the treatment of various ailments. Two of the most prominent tyrosine kinases as drug targets are the epidermal growth factor receptor (EGFR) and the vascular endothelial growth factor receptor (VEGFR), as evidenced by the clinical success of their many inhibitors in the drug market. Among several other physiological roles, EGFR regulates epithelial tissue development and homeostasis, while VEGFR regulates tumor-induced angiogenesis. The pyrrolo[2,3-d]pyrimidine nucleus represents a deaza-isostere of adenine, the nitrogenous base of ATP. The recent introduction of many pyrrolo[2,3-d]pyrimidines to the drug market as tyrosine kinase inhibitors makes them a hot topic in the medicinal chemistry research area at the present time. This review article comprehensively sheds light on the structure-activity relationship (SAR) of pyrrolo[2,3-d]pyrimidines as EGFR and VEGFR tyrosine kinase inhibitors, aiming to provide help medicinal chemists in the design of future pyrrolopyrimidine kinase inhibitors.
Collapse
Affiliation(s)
- Kamel Metwally
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk 71491, Saudi Arabia
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk 71491, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
33
|
Zhou H, Fu H, Shao X, Cai W. Binding Thermodynamics of Fourth-Generation EGFR Inhibitors Revealed by Absolute Binding Free Energy Calculations. J Chem Inf Model 2023; 63:7837-7846. [PMID: 38054791 DOI: 10.1021/acs.jcim.3c01636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
The overexpression or mutation of the kinase domain of the epidermal growth factor receptor (EGFR) is strongly associated with non-small-cell lung cancer (NSCLC). EGFR tyrosine kinase inhibitors (TKIs) have proven to be effective in treating NSCLC patients. However, EGFR mutations can result in drug resistance. To elucidate the mechanisms underlying this resistance and inform future drug development, we examined the binding affinities of BLU-945, a recently reported fourth-generation TKI, to wild-type EGFR (EGFRWT) and its double-mutant (L858R/T790M; EGFRDM) and triple-mutant (L858R/T790M/C797S; EGFRTM) forms. We compared the binding affinities of BLU-945, BLU-945 analogues, CH7233163 (another fourth-generation TKI), and erlotinib (a first-generation TKI) using absolute binding free energy calculations. Our findings reveal that BLU-945 and CH7233163 exhibit binding affinities to both EGFRDM and EGFRTM stronger than those of erlotinib, corroborating experimental data. We identified K745 and T854 as the key residues in the binding of fourth-generation EGFR TKIs. Electrostatic forces were the predominant driving force for the binding of fourth-generation TKIs to EGFR mutants. Furthermore, we discovered that the incorporation of piperidinol and sulfone groups in BLU-945 substantially enhanced its binding capacity to EGFR mutants. Our study offers valuable theoretical insights for optimizing fourth-generation EGFR TKIs.
Collapse
Affiliation(s)
- Huaxin Zhou
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| |
Collapse
|
34
|
Guo T, Wu C, Zhang J, Yu J, Li G, Jiang H, Zhang X, Yu R, Liu X. Dual blockade of EGFR and PI3K signaling pathways offers a therapeutic strategy for glioblastoma. Cell Commun Signal 2023; 21:363. [PMID: 38115126 PMCID: PMC10729576 DOI: 10.1186/s12964-023-01400-0] [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: 08/03/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a devastating disease that lacks effective drugs for targeted therapy. Previously, we found that the third-generation epidermal growth factor receptor (EGFR) inhibitor AZD-9291 persistently blocked the activation of the ERK pathway but had no inhibitory effect on the phosphoinositide 3-kinase (PI3K)/Akt pathway. Given that the PI3K inhibitor GDC-0084 is being evaluated in phase I/II clinical trials of GBM treatment, we hypothesized that combined inhibition of the EGFR/ERK and PI3K/Akt pathways may have a synergistic effect in the treatment of GBM. METHODS The synergistic effects of cotreatment with AZD-9291 and GDC-0084 were validated using cell viability assays in GBM and primary GBM cell lines. Moreover, the underlying inhibitory mechanisms were assessed through colony formation, EdU proliferation, and cell cycle assays, as well as RNA-seq analyses and western blot. The therapeutic effects of the drug combination on tumor growth and survival were investigated in mice bearing tumors using subcutaneously or intracranially injected LN229 xenografts. RESULTS Combined treatment with AZD-9291 and GDC-0084 synergistically inhibited the proliferation and clonogenic survival, as well as induced cell cycle arrest of GBM cells and primary GBM cells, compared to monotherapy. Moreover, AZD-9291 plus GDC-0084 combination therapy significantly inhibited the growth of subcutaneous tumors and orthotopic brain tumor xenografts, thus prolonging the survival of tumor-bearing mice. More importantly, the combination of AZD-9291 and GDC-0084 simultaneously blocked the activation of the EGFR/MEK/ERK and PI3K/AKT/mTOR signaling pathways, thereby exerting significant antitumor activity. CONCLUSION Our findings demonstrate that the combined blockade of the EGFR/MEK/ERK and PI3K/AKT/mTOR pathways is more effective against GBM than inhibition of each pathway alone, both in vitro and in vivo. Our results suggest that AZD-9291 combined with GDC-0084 may be considered as a potential treatment strategy in future clinical trials. Video Abstract.
Collapse
Affiliation(s)
- Tongxuan Guo
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Changyong Wu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junhao Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiefeng Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guoxi Li
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongyan Jiang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
35
|
Niu ZX, Wang YT, Lu N, Sun JF, Nie P, Herdewijn P. Advances of clinically approved small-molecule drugs for the treatment of non-small cell lung cancer. Eur J Med Chem 2023; 261:115868. [PMID: 37844346 DOI: 10.1016/j.ejmech.2023.115868] [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: 07/25/2023] [Revised: 09/14/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Lung cancer continues to pose a significant challenge as a prominent contributor to global cancer-related mortality. Despite the considerable strides made in therapeutic interventions within the past decade, a substantial population of patients diagnosed with non-small cell lung cancer (NSCLC) still face the grim reality of an incurable condition. In the realm of optimal management strategies for individuals afflicted with locally advanced, yet amenable to surgical resection, NSCLC, a therapeutic approach encompassing chemoradiation stands as a fundamental component. Significant strides have been made in the therapeutic landscape of NSCLC during the preceding two decades, facilitating an enhanced comprehension of the underlying disease biology, and mechanisms governing tumor progression, as well as advancements in early detection modalities and multimodal therapeutic interventions. Nevertheless, the overall rates of curative interventions and survival outcomes for NSCLC continue to exhibit a discouragingly low trajectory, particularly in the context of metastatic disease. Hence, the imperative for sustained research endeavors in the realm of novel pharmaceutical agents and combinatorial therapeutic approaches remains paramount, with the overarching objective of broadening the scope of clinical advantages conferred upon a wider demographic of patients, thereby fostering tangible improvements in outcomes pertaining to NSCLC. The primary objective of this review is to provide an all-encompassing examination encompassing the clinical application and synthetic routes of specific drugs, with the explicit aim of disseminating invaluable knowledge that can inform future research and development endeavors focused on NSCLC.
Collapse
Affiliation(s)
- Zhen-Xi Niu
- Department of Pharmacy, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Ya-Tao Wang
- Department of Pharmacy, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China; First People's Hospital of Shangqiu, Henan Province, Shangqiu, 476100, China
| | - Nan Lu
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| | - Jin-Feng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin, 133002, China.
| | - Peng Nie
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| |
Collapse
|
36
|
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: 1] [Impact Index Per Article: 0.5] [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
|
37
|
Zhou Y, Huang S, Guo Y, Ran M, Shan W, Chen WH, Tam KY. Epigallocatechin gallate circumvents drug-induced resistance in non-small-cell lung cancer by modulating glucose metabolism and AMPK/AKT/MAPK axis. Phytother Res 2023; 37:5837-5853. [PMID: 37621136 DOI: 10.1002/ptr.7990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/25/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
Upon prolonged use of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancer (NSCLC), acquired drug resistance inevitably occurs. This study investigates the combined use of EGFR-TKIs (gefitinib or osimertinib) with epigallocatechin gallate (EGCG) to overcome acquired drug resistance in NSCLC models. The in vitro antiproliferative effects of EGFR-TKIs and EGCG combination in EGFR-mutant parental and resistant cell lines were evaluated. The in vivo efficacy of the combination was assessed in xenograft mouse models derived from EGFR-TKI-resistant NSCLC cells. We found that the combined use of EGFR-TKIs and EGCG significantly reversed the Warburg effect by suppressing glycolysis while boosting mitochondrial respiration, which was accompanied by increased cellular ROS and decreased lactate secretion. The combination effectively activated the AMPK pathway while inhibited both ERK/MAPK and AKT/mTOR pathways, leading to cell cycle arrest and apoptosis, particularly in drug-resistant NSCLC cells. The in vivo results obtained from mouse tumor xenograft model confirmed that EGCG effectively overcame osimertinib resistance. This study revealed that EGCG suppressed cancer bypass survival signaling and altered cancer metabolic profiles, which is a promising anticancer adjuvant of EGFR-TKIs to overcome acquired drug resistance in NSCLC.
Collapse
Affiliation(s)
- Yan Zhou
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Shiqi Huang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Yizhen Guo
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Maoxin Ran
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Wenying Shan
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Wen-Hua Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| |
Collapse
|
38
|
Abo Al-Hamd MG, Tawfik HO, Abdullah O, Yamaguchi K, Sugiura M, Mehany ABM, El-Hamamsy MH, El-Moselhy TF. Recruitment of hexahydroquinoline as anticancer scaffold targeting inhibition of wild and mutants EGFR (EGFR WT, EGFR T790M, and EGFR L858R). J Enzyme Inhib Med Chem 2023; 38:2241674. [PMID: 37548154 PMCID: PMC10408569 DOI: 10.1080/14756366.2023.2241674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/14/2023] [Accepted: 07/23/2023] [Indexed: 08/08/2023] Open
Abstract
Hexahydroquinoline (HHQ) scaffold was constructed and recruited for development of new series of anticancer agents. Thirty-two new compounds were synthesised where x-ray crystallography was performed to confirm enantiomerism. Thirteen compounds showed moderate to good activity against NCI 60 cancer cell lines, with GI % mean up to 74% for 10c. Expending erlotinib as a reference drug, target compounds were verified for their inhibiting activities against EGFRWT, EGFRT790M, and EGFRL858R where compound 10d was the best inhibitor with IC50 = 0.097, 0.280, and 0.051 µM, respectively, compared to erlotinib (IC50 = 0.082 µM, 0.342 µM, and 0.055 µM, respectively). Safety profile was validated using normal human lung (IMR-90) cells. 10c and 10d disrupted cell cycle at pre-G1 and G2/M phases in lung cancer, HOP-92, and cell line. Molecular docking study was achieved to understand the potential binding interactions and affinities in the active sites of three versions of EGFRs.
Collapse
Affiliation(s)
- Mahmoud G. Abo Al-Hamd
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Haytham O. Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Omeima Abdullah
- Pharmaceutical Chemistry Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Koki Yamaguchi
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Masaharu Sugiura
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Ahmed B. M. Mehany
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mervat H. El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Tarek F. El-Moselhy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
39
|
Jin F, Ni X, Yu S, Jiang X, Zhou J, Mao D, Liu Y, Wu F. Network pharmacology‑based investigation of potential targets of triptonodiol acting on non-small-cell lung cancer. Eur J Med Res 2023; 28:547. [PMID: 38017514 PMCID: PMC10683219 DOI: 10.1186/s40001-023-01453-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/17/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Triptonodiol is a very promising antitumor drug candidate extracted from the Chinese herbal remedy Tripterygium wilfordii Hook. F., and related studies are underway. METHODS To explore the mechanism of triptonodiol for lung cancer treatment, we used network pharmacology, molecular docking, and ultimately protein validation. Gene ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis were performed through the David database. Molecular docking was performed using PyMoL2.3.0 and AutoDock Vina software. After screening, the major targets of triptonodiol were identified for the treatment of lung cancer. Target networks were established, Protein-protein interaction (PPI) network topology was analyzed, then KEGG pathway enrichment analysis was performed. Useful proteins were screened by survival analysis, and Western blot analysis was performed. RESULTS Triptonodiol may regulate cell proliferation, drug resistance, metastasis, anti-apoptosis, etc., by acting on glycogen synthase kinase 3 beta (GSK3B), protein kinase C (PKC), p21-activated kinase (PAK), and other processes. KEGG pathway enrichment analysis showed that these targets were associated with tumor, erythroblastic oncogene B (ErbB) signaling, protein phosphorylation, kinase activity, etc. Molecular docking showed that the target protein GSK has good binding activity to the main active component of triptonodiol. The protein abundance of GSK3B was significantly downregulated in non-small-cell lung cancer cells H1299 and A549 treated with triptonodiol for 24 h. CONCLUSION The cellular-level studies combined with network pharmacology and molecular docking approaches provide new ideas for the development and therapeutic application of triptonodiol, and identify it as a potential GSK inhibitor.
Collapse
Affiliation(s)
- Feng Jin
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Xiaochen Ni
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Shilong Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou, 225001, People's Republic of China
| | - Xiaomin Jiang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, People's Republic of China
| | - Jun Zhou
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Defang Mao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, People's Republic of China
| | - Feng Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
40
|
Tang G, Wang W, Wang X, Ding K, Ngan SC, Chen JY, Sze SK, Gao L, Yuan P, Lu X, Yao SQ. Cell-active, irreversible covalent inhibitors that selectively target the catalytic lysine of EGFR by using fluorosulfate-based SuFEx chemistry. Eur J Med Chem 2023; 259:115671. [PMID: 37499291 DOI: 10.1016/j.ejmech.2023.115671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
EGFR signaling is involved in multiple cellular processes including cell proliferation, differentiation and development, making this protein kinase one of the most valuable drug targets for the treatment of non-small cell lung carcinomas (NSCLC). Herein, we describe the design and synthesis of a series of potential covalent inhibitors targeting the catalytically conserved lysine (K745) of EGFR on the basis of Erlotinib, an FDA-approved first-generation EGFR drug. Different amine-reactive electrophiles were introduced at positions on the Erlotinib scaffold proximal to K745 in EGFR. The optimized compound 26 (as well as its close analog 30), possessing a novel arylfluorosulfate group (ArOSO2F), showed excellent in vitro potency (as low as 0.19 nM in independent IC50 determination) and selectivity against EGFR and many of its drug-resistant mutants. Both intact protein mass spectrometry (MS) and site-mapping analysis revealed that compound 26 covalently bound to EGFR at K745 through the formation of a sulfamate. In addition, compound 26 displayed good anti-proliferative potency against EGFR-overexpressing HCC827 cells by inhibiting endogenous EGFR autophosphorylation. The pharmacokinetic studies of compound 26 demonstrated the druggable potential of other ArOSO2F-containing compounds. Finally, competitive activity-based protein profiling (ABPP), cellular thermal shift assay (CETSA), as well as cellular wash-out experiments, all showed compound 26 to be the first cell-active, fluorosulfate-based targeted covalent inhibitor (TCI) of protein kinases capable of covalently engaging the catalytically conserved lysine of its target in live mammalian cells.
Collapse
Affiliation(s)
- Guanghui Tang
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Wei Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518000, China
| | - Xuan Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518000, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, 510632, China; State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - SoFong Cam Ngan
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Jiao-Yu Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518000, China
| | - Siu Kwan Sze
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518000, China
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518000, China.
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore.
| |
Collapse
|
41
|
Singh S, Sadhukhan S, Sonawane A. 20 years since the approval of first EGFR-TKI, gefitinib: Insight and foresight. Biochim Biophys Acta Rev Cancer 2023; 1878:188967. [PMID: 37657684 DOI: 10.1016/j.bbcan.2023.188967] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/14/2023] [Accepted: 08/20/2023] [Indexed: 09/03/2023]
Abstract
Epidermal growth factor receptor (EGFR) actively involves in modulation of various cancer progression related mechanisms including angiogenesis, differentiation and migration. Therefore, targeting EGFR has surfaced as a prominent approach for the treatment of several types of cancers, including non-small cell lung cancer (NSCLC), pancreatic cancer, glioblastoma. Various first, second and third generation of EGFR tyrosine kinase inhibitors (EGFR-TKIs) have demonstrated effectiveness as an anti-cancer therapeutics. However, rapid development of drug resistance and mutations still remains a major challenge for the EGFR-TKIs therapy. Overcoming from intrinsic and acquired resistance caused by EGFR mutations warrants the further exploration of alternative strategies and discovery of novel inhibitors. In this review, we delve into the breakthrough discoveries have been made in previous 20 years, and discuss the currently ongoing efforts aimed to circumvent the chemo-resistance. We also highlight the new challenges, limitations and future directions for the development of improved therapeutic approaches such as fourth-generation EGFR-TKIs, peptides, nanobodies, PROTACs etc.
Collapse
Affiliation(s)
- Satyam Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Madhya Pradesh 453 552, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala 678 623, India; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Kerala 678 623, India.
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Madhya Pradesh 453 552, India.
| |
Collapse
|
42
|
Fan Y, Wang X, Dong Y, Cui E, Wang H, Sun X, Su J, Luo Y, Yu T, Jiang X. Multiregional radiomics of brain metastasis can predict response to EGFR-TKI in metastatic NSCLC. Eur Radiol 2023; 33:7902-7912. [PMID: 37142868 DOI: 10.1007/s00330-023-09709-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/12/2023] [Accepted: 03/16/2023] [Indexed: 05/06/2023]
Abstract
OBJECTIVES To develop radiomics signatures from multiparametric magnetic resonance imaging (MRI) scans to detect epidermal growth factor receptor (EGFR) mutations and predict the response to EGFR-tyrosine kinase inhibitors (EGFR-TKIs) in non-small cell lung cancer (NSCLC) patients with brain metastasis (BM). METHODS We included 230 NSCLC patients with BM treated at our hospital between January 2017 and December 2021 and 80 patients treated at another hospital between July 2014 and October 2021 to form the primary and external validation cohorts, respectively. All patients underwent contrast-enhanced T1-weighted (T1C) and T2-weighted (T2W) MRI, and radiomics features were extracted from both the tumor active area (TAA) and peritumoral edema area (POA) for each patient. The least absolute shrinkage and selection operator (LASSO) was used to identify the most predictive features. Radiomics signatures (RSs) were constructed using logistic regression analysis. RESULTS For predicting the EGFR mutation status, the created RS-EGFR-TAA and RS-EGFR- POA showed similar performance. By combination of TAA and POA, the multi-region combined RS (RS-EGFR-Com) achieved the highest prediction performance, with AUCs of 0.896, 0.856, and 0.889 in the primary training, internal validation, and external validation cohort, respectively. For predicting response to EGFR-TKI, the multi-region combined RS (RS-TKI-Com) generated the highest AUCs in the primary training (AUC = 0.817), internal validation (AUC = 0.788), and external validation (AUC = 0.808) cohort, respectively. CONCLUSIONS Our findings suggested values of multiregional radiomics of BM for predicting EGFR mutations and response to EGFR-TKI. CLINICAL RELEVANCE STATEMENT The application of radiomic analysis of multiparametric brain MRI has proven to be a promising tool to stratify which patients can benefit from EGFR-TKI therapy and to facilitate the precise therapeutics of NSCLC patients with brain metastases. KEY POINTS • Multiregional radiomics can improve efficacy in predicting therapeutic response to EGFR-TKI therapy in NSCLC patients with brain metastasis. • The tumor active area (TAA) and peritumoral edema area (POA) may hold complementary information related to the therapeutic response to EGFR-TKI. • The developed multi-region combined radiomics signature achieved the best predictive performance and may be considered as a potential tool for predicting response to EGFR-TKI.
Collapse
Affiliation(s)
- Ying Fan
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xinti Wang
- The First Clinical Department, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yue Dong
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Enuo Cui
- School of Computer Science and Engineering, Shenyang University, Shenyang, 110044, People's Republic of China
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Xinyan Sun
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Juan Su
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Tao Yu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| | - Xiran Jiang
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
43
|
Sakharkar P, Kurup S. Comparing Efficacy of Erlotinib and Bevacizumab Combination with Erlotinib Monotherapy in Patients with Advanced Non-Small Cell Lung Cancer (NSCLC): A Systematic Review and Meta-Analysis. Diseases 2023; 11:146. [PMID: 37873790 PMCID: PMC10594499 DOI: 10.3390/diseases11040146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/24/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023] Open
Abstract
The objective of this systematic review and meta-analysis was to assess and contrast the efficacy and safety of combining erlotinib and bevacizumab with erlotinib alone in the treatment of patients with advanced non-small cell lung cancer (NSCLC). The authors searched databases such as PubMed, Medline, Scopus, and Cochrane Central Register of Controlled Trials for randomized control trials (RCTs) comparing erlotinib plus bevacizumab with erlotinib in NSCLC patients. The overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs) were the outcomes of interest. The pooled hazard ratio (HR) and relative risk (RR) were estimated utilizing both fixed- and random-effect models. Methodological quality of the included studies was assessed using the Cochrane Risk of Bias tool. Nine studies comprising 1698 patients with NSCLC were included in this meta-analysis, of whom 850 were treated with erlotinib plus bevacizumab, and 848 with erlotinib. The erlotinib plus bevacizumab combination significantly prolonged PFS (HR, 0.62, 95% CI: 0.56, 0.70, p < 0.00001) but did not show any significant improvement in OS (HR, 0.95; 95% CI: 0.83, 1.07, p = 0.39) and ORR (HR, 1.10; 95% CI: 0.98, 1.24, p = 0.09). Increased risks of hypertension (RR, 5.15; 95% CI: 3.59, 7.39; p < 0.00001), proteinuria (RR, 10.54; 95% CI: 3.80, 29.20; p < 0.00001) and grade 3 and higher AEs (RR, 2.09; 95% CI: 1.47, 2.97; p < 0.00001) were observed with the erlotinib-plus-bevacizumab combination compared to erlotinib monotherapy. On subgroup analyses, the erlotinib plus bevacizumab combination improved PFS only. Combining erlotinib and bevacizumab has been shown to improve PFS in advanced NSCLC patients but did not show any significant OS and ORR benefits. Furthermore, risks of hypertension, proteinuria, and grade 3 or higher AEs were greater with the erlotinib-and-bevacizumab combination.
Collapse
Affiliation(s)
- Prashant Sakharkar
- Department of Clinical and Administrative Sciences, College of Science, Health and Pharmacy, Roosevelt University, Schaumburg, IL 60173, USA
| | - Sonali Kurup
- Department of Pharmaceutical Sciences, College of Pharmacy, Ferris State University, Big Rapids, MI 49307, USA;
| |
Collapse
|
44
|
Al-Wahaibi LH, Abou-Zied HA, Hisham M, Beshr EAM, Youssif BGM, Bräse S, Hayallah AM, Abdel-Aziz M. Design, Synthesis, and Biological Evaluation of Novel 3-Cyanopyridone/Pyrazoline Hybrids as Potential Apoptotic Antiproliferative Agents Targeting EGFR/BRAF V600E Inhibitory Pathways. Molecules 2023; 28:6586. [PMID: 37764362 PMCID: PMC10537368 DOI: 10.3390/molecules28186586] [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: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
A series of novel 3-cyanopyridone/pyrazoline hybrids (21-30) exhibiting dual inhibition against EGFR and BRAFV600E has been developed. The synthesized target compounds were tested in vitro against four cancer cell lines. Compounds 28 and 30 demonstrated remarkable antiproliferative activity, boasting GI50 values of 27 nM and 25 nM, respectively. These hybrids exhibited dual inhibitory effects on both EGFR and BRAFV600E pathways. Compounds 28 and 30, akin to Erlotinib, displayed promising anticancer potential. Compound 30 emerged as the most potent inhibitor against cancer cell proliferation and BRAFV600E. Notably, both compounds 28 and 30 induced apoptosis by elevating levels of caspase-3 and -8 and Bax, while downregulating the antiapoptotic Bcl2 protein. Molecular docking studies confirmed the potential of compounds 28 and 30 to act as dual EGFR/BRAFV600E inhibitors. Furthermore, in silico ADMET prediction indicated that most synthesized 3-cyanopyridone/pyrazoline hybrids exhibit low toxicity and minimal adverse effects.
Collapse
Affiliation(s)
- Lamya H. Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11564, Saudi Arabia;
| | - Hesham A. Abou-Zied
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt; (H.A.A.-Z.); (M.H.)
| | - Mohamed Hisham
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt; (H.A.A.-Z.); (M.H.)
| | - Eman A. M. Beshr
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (E.A.M.B.); (M.A.-A.)
| | - Bahaa G. M. Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Alaa M. Hayallah
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Sphinx University, Assiut 71515, Egypt
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (E.A.M.B.); (M.A.-A.)
| |
Collapse
|
45
|
Li F, Wu H, Du X, Sun Y, Rausseo BN, Talukder A, Katailiha A, Elzohary L, Wang Y, Wang Z, Lizée G. Epidermal Growth Factor Receptor-Targeted Neoantigen Peptide Vaccination for the Treatment of Non-Small Cell Lung Cancer and Glioblastoma. Vaccines (Basel) 2023; 11:1460. [PMID: 37766136 PMCID: PMC10534925 DOI: 10.3390/vaccines11091460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) plays crucial roles in several important biological functions such as embryogenesis, epithelial tissue development, and cellular regeneration. However, in multiple solid tumor types overexpression and/or activating mutations of the EGFR gene frequently occur, thus hijacking the EGFR signaling pathway to promote tumorigenesis. Non-small cell lung cancer (NSCLC) tumors in particular often contain prevalent and shared EGFR mutations that provide an ideal source for public neoantigens (NeoAg). Studies in both humans and animal models have confirmed the immunogenicity of some of these NeoAg peptides, suggesting that they may constitute viable targets for cancer immunotherapies. Peptide vaccines targeting mutated EGFR have been tested in multiple clinical trials, demonstrating an excellent safety profile and encouraging clinical efficacy. For example, the CDX-110 (rindopepimut) NeoAg peptide vaccine derived from the EGFRvIII deletion mutant in combination with temozolomide and radiotherapy has shown efficacy in treating EGFRvIII-harboring glioblastoma multiforme (GBM) patients undergone surgery in multiple Phase I and II clinical trials. Furthermore, pilot clinical trials that have administered personalized NeoAg peptides for treating advanced-stage NSCLC patients have shown this approach to be a feasible and safe method to increase antitumor immune responses. Amongst the vaccine peptides administered, EGFR mutation-targeting NeoAgs induced the strongest T cell-mediated immune responses in patients and were also associated with objective clinical responses, implying a promising future for NeoAg peptide vaccines for treating NSCLC patients with selected EGFR mutations. The efficacy of NeoAg-targeting peptide vaccines may be further improved by combining with other modalities such as tyrosine kinase or immune checkpoint inhibitor (ICI) therapy, which are currently being tested in animal models and clinical trials. Herein, we review the most current basic and clinical research progress on EGFR-targeted peptide vaccination for the treatment of NSCLC and other solid tumor types.
Collapse
Affiliation(s)
- Fenge Li
- Core Laboratory, Tianjin Beichen Hospital, Tianjin 300400, China
- Department of Oncology, Tianjin Beichen Hospital, Tianjin 300400, China
| | - Huancheng Wu
- Department of Neurosurgery, Tianjin Beichen Hospital, Tianjin 300400, China
| | - Xueming Du
- Department of Oncology, Tianjin Beichen Hospital, Tianjin 300400, China
| | - Yimo Sun
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Barbara Nassif Rausseo
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Amjad Talukder
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Arjun Katailiha
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Lama Elzohary
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Yupeng Wang
- Department of Oncology, Tianjin Beichen Hospital, Tianjin 300400, China
| | - Zhiyu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Gregory Lizée
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
46
|
Urbanska EM, Grauslund M, Koffeldt PR, Truelsen SLB, Löfgren JO, Costa JC, Melchior LC, Sørensen JB, Santoni-Rugiu E. Real-World Data on Combined EGFR-TKI and Crizotinib Treatment for Acquired and De Novo MET Amplification in Patients with Metastatic EGFR-Mutated NSCLC. Int J Mol Sci 2023; 24:13077. [PMID: 37685884 PMCID: PMC10487649 DOI: 10.3390/ijms241713077] [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: 07/30/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Amplification of the mesenchymal epithelial transition (MET) gene is a mechanism of acquired resistance to epidermal growth factor receptor (EGFR)-tyrosine-kinase-inhibitors (TKIs) in over 20% of patients with advanced EGFR-mutated (EGFRm+) non-small lung cancer (NSCLC). However, it may also occur de novo in 2-8% of EGFRm+ NSCLC cases as a potential mechanism of intrinsic resistance. These patients represent a group with unmet needs, since there is no standard therapy currently approved. Several new MET inhibitors are being investigated in clinical trials, but the results are awaited. Meanwhile, as an alternative strategy, combinations of EGFR-TKIs with the MET/ALK/ROS1-TKI Crizotinib may be used in this setting, despite this use is principally off-label. Thus, we studied five of these MET amplified cases receiving EGFR-TKI and Crizotinib doublet after progression on EGFR-TKI treatment to assess the benefits and challenges related to this combination and the possible occurrence of genomic and phenotypic co-alterations. Furthermore, we compared our cases with other real-world reports on Crizotinib/EGFR-TKI combinations, which appeared effective, especially in patients with high-level MET amplification. Yet, we observed that the co-occurrence of other genomic and phenotypical alterations may affect the response to combined EGFR-TKI and Crizotinib. Finally, given the heterogeneity of MET amplification, the diagnostic methods for assessing it may be discrepant. In this respect, we observed that for optimal detection, immunohistochemistry, fluorescence in situ hybridization, and next-generation sequencing should be used together, as these methods possess different sensitivities and complement each other in characterizing MET amplification. Additionally, we addressed the issue of managing EGFR-mutated NSCLC patients with de novo MET amplification causing primary EGFR-TKI resistance. We conclude that, while data from clinical trials with new MET inhibitors are still pending, adding Crizotinib to EGFR-TKI in NSCLC patients acquiring MET amplification at progression on EGFR-TKI monotherapy is a reasonable approach, with a progression-free survival of 3-19 months.
Collapse
Affiliation(s)
- Edyta M. Urbanska
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
| | - Morten Grauslund
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Peter R. Koffeldt
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Sarah L. B. Truelsen
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Johan O. Löfgren
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
| | - Junia C. Costa
- Department of Radiology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
| | - Linea C. Melchior
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
| | - Jens B. Sørensen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Eric Santoni-Rugiu
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; (M.G.); (P.R.K.); (S.L.B.T.); (L.C.M.)
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
47
|
Chen R, Hao X, Chen J, Zhang C, Fan H, Lian F, Chen X, Wang C, Xia Y. Integrated multi-omics analyses reveal Jorunnamycin A as a novel suppressor for muscle-invasive bladder cancer by targeting FASN and TOP1. J Transl Med 2023; 21:549. [PMID: 37587470 PMCID: PMC10428641 DOI: 10.1186/s12967-023-04400-3] [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: 03/10/2023] [Accepted: 07/29/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Bladder cancer is a urological carcinoma with high incidence, among which muscle invasive bladder cancer (MIBC) is a malignant carcinoma with high mortality. There is an urgent need to develop new drugs with low toxicity and high efficiency for MIBC because existing medication has defects, such as high toxicity, poor efficacy, and side effects. Jorunnamycin A (JorA), a natural marine compound, has been found to have a high efficiency anticancer effect, but its anticancer function and mechanism on bladder cancer have not been studied. METHODS To examine the anticancer effect of JorA on MIBC, Cell Counting Kit 8, EdU staining, and colony formation analyses were performed. Moreover, a xenograft mouse model was used to verify the anticancer effect in vivo. To investigate the pharmacological mechanism of JorA, high-throughput quantitative proteomics, transcriptomics, RT-qPCR, western blotting, immunofluorescence staining, flow cytometry, pulldown assays, and molecular docking were performed. RESULTS JorA inhibited the proliferation of MIBC cells, and the IC50 of T24 and UM-UC-3 was 0.054 and 0.084 μM, respectively. JorA-induced significantly changed proteins were enriched in "cancer-related pathways" and "EGFR-related signaling pathways", which mainly manifested by inhibiting cell proliferation and promoting cell apoptosis. Specifically, JorA dampened the DNA synthesis rate, induced phosphatidylserine eversion, and inhibited cell migration. Furthermore, it was discovered that fatty acid synthase (FASN) and topoisomerase 1 (TOP1) are the JorA interaction proteins. Using DockThor software, the 3D docking structures of JorA binding to FASN and TOP1 were obtained (the binding affinities were - 8.153 and - 7.264 kcal/mol, respectively). CONCLUSIONS The marine compound JorA was discovered to have a specific inhibitory effect on MIBC, and its potential pharmacological mechanism was revealed for the first time. This discovery makes an important contribution to the development of new high efficiency and low toxicity drugs for bladder cancer therapy.
Collapse
Affiliation(s)
- Ruijiao Chen
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining, 272067, Shandong, China
| | - Xiaopeng Hao
- Institute of Precision Medicine, Jining Medical University, No. 133 Hehua Road, Taibaihu District, Jining, 272067, Shandong, China
| | - Jingyuan Chen
- Institute of Precision Medicine, Jining Medical University, No. 133 Hehua Road, Taibaihu District, Jining, 272067, Shandong, China
| | - Changyue Zhang
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining, 272067, Shandong, China
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Huixia Fan
- Institute of Precision Medicine, Jining Medical University, No. 133 Hehua Road, Taibaihu District, Jining, 272067, Shandong, China
| | - Fuming Lian
- Institute of Precision Medicine, Jining Medical University, No. 133 Hehua Road, Taibaihu District, Jining, 272067, Shandong, China
| | - Xiaochuan Chen
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Chao Wang
- Department of Urology, Jining No. 1 People's Hospital, Jining, 272106, Shandong, China.
| | - Yong Xia
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining, 272067, Shandong, China.
- Institute of Precision Medicine, Jining Medical University, No. 133 Hehua Road, Taibaihu District, Jining, 272067, Shandong, China.
| |
Collapse
|
48
|
Lu H, Zhang B, Xie Y, Zhao W, Han W, Zhou L, Wang Z. Sitravatinib is a potential EGFR inhibitor and induce a new death phenotype in Glioblastoma. Invest New Drugs 2023; 41:564-578. [PMID: 37322389 DOI: 10.1007/s10637-023-01373-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023]
Abstract
Glioblastoma (GBM) is a highly lethal neurological tumor that presents significant challenge for clinicians due to its heterogeneity and high mortality rate. Despite extensive research, there is currently no effective drug treatment available for GBM. Research evidence has consistently demonstrated that the epidermal growth factor receptor (EGFR) promotes tumor progression and is associated with poor prognosis in several types of cancer. In glioma, EGFR abnormal amplification is reported in approximately 40% of GBM patients, with overexpression observed in 60% of cases, and deletion or mutation in 24% to 67% of patients. In our study, Sitravatinib, a potential EGFR inhibitor, was identified through molecular docking screening based on protein structure. The targeting of EGFR and the tumor inhibitory effect of Sitravatinib on glioma were verified through cellular and in vivo experiments, respectively. Our study also revealed that Sitravatinib effectively inhibited GBM invasive and induced DNA damage and cellular senescence. Furthermore, we observed a novel cell death phenotype induced by Sitravatinib, which differed from previously reported programmed death patterns such as apoptosis, pyroptosis, ferroptosis, and necrosis.
Collapse
Affiliation(s)
- Hanwen Lu
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen City, China
| | - Bingchang Zhang
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen City, China
| | - Yuanyuan Xie
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen City, China
| | - Wenpeng Zhao
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen City, China
| | - Wanhong Han
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen City, China
| | - Liwei Zhou
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen City, China
| | - Zhanxiang Wang
- The Department of Neuroscience, Institute of Neurosurgery, School of Medicine, Xiamen University, Xiamen City, China.
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen City, China.
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen City, China.
- Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen City, China.
| |
Collapse
|
49
|
Zhou J, Chu X, Zhao J, Xie M, Wu J, Yu X, Fang Y, Li Y, Li X, Su C. Full spectrum flow cytometry-powered comprehensive analysis of PBMC as biomarkers for immunotherapy in NSCLC with EGFR-TKI resistance. Biol Proced Online 2023; 25:21. [PMID: 37488517 PMCID: PMC10364374 DOI: 10.1186/s12575-023-00215-0] [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: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Clinical studies suggest that immune checkpoint inhibitor (ICI) monotherapy has limited benefits in non-small cell lung cancer (NSCLC) patients after epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) failure. However, data about efficacy of ICI plus chemotherapy remain controversial, probably attributed to the heterogeneity among such population, and robust efficacy biomarkers are urgent to explore. METHODS A total of 60 eligible patients who received ICI plus chemotherapy after EGFR-TKI treatment failure were enrolled, 24 of whom peripheral blood mononuclear cell (PBMC) samples were collected at baseline and after 2 cycles of treatment. We have designed a 23-color-antibody panel to detect PBMC by full spectrum flow cytometry. RESULTS For EGFR-TKI resistant NSCLC patients: 1) ICI plus chemotherapy achieved an objective response rate (ORR) of 21.7% and a median progression-free survival (PFS) of 6.4 months. 2) clinical characteristics associated with worse efficacy included liver metastasis and platelet-to-lymphocyte ratio (PLR) > 200. 3) the proportion of immune cell subset associated with better efficacy was higher baseline effective CD4+T cells (E4). 4) the baseline expression of immune checkpoint proteins (ICPs) on cell subsets associated with better efficacy included: higher expression of CD25 on dendritic cells (DC) and central memory CD8+T cells (CM8), and higher expression of Lymphocyte activation gene 3 (LAG-3) on effective memory CD8+T cells (EM8). 5) the expression of ICPs after 2 cycles of treatment associated with better efficacy included: higher expression of CD25 on CD8+T/EM8 /natural killer (NK) cells. 6) the dynamic changes of ICPs expression associated with worse efficacy included: significantly decrease of T cell immunoglobulin and ITIM domain (TIGIT) expression on regular T cells (Tregs) and decrease of V-domain immunoglobulin suppressor of T cell activation (VISTA) expression on Th1. 7) a prediction model for the efficacy of ICI plus chemotherapy was successfully constructed with a sensitivity of 62.5%, specificity of 100%, and area under curve (AUC) = 0.817. CONCLUSIONS Some EGFR-TKI-resistant NSCLC patients could indeed benefit from ICI plus chemotherapy, but most patients are primary resistant to immunotherapy. Comprehensive analysis of peripheral immune cells using full spectrum flow cytometry showed that compared to the proportion of cell subsets, the expression type and level of ICPs on immune cells, especially CD25, were significantly correlated with the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Xiangling Chu
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Jing Zhao
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Mengqing Xie
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Jing Wu
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Xin Yu
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Yujia Fang
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Yazhou Li
- Righton Biotechnology Co., Ltd, Shanghai, China
| | - Xiyan Li
- Righton Biotechnology Co., Ltd, Shanghai, China
| | - Chunxia Su
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China.
| |
Collapse
|
50
|
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
|