1
|
Yu Q, Peng X, Xu G, Bai X, Cao Y, Du Y, Wang X, Zhao R. Overexpression or knockdown of the P2X7 receptor regulates the progression of non-small cell lung cancer, involving GSK-3β and JNK signaling pathways. Eur J Pharmacol 2025; 995:177421. [PMID: 39993700 DOI: 10.1016/j.ejphar.2025.177421] [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: 10/11/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
Studies have indicated that P2X7 receptor are involved in the progression of non-small cell lung cancer (NSCLC). Therefore, this study sought to explore how modulating P2X7 receptor expression levels affect the biological function of NSCLC and its underlying mechanisms. Recombinant plasmids with P2X7 receptor overexpression or knockdown were constructed and transfected into LLC and LA795 cells, and the biological function changes of these two cells were assessed in vitro. Subsequently, stable cell lines (overexpression or knockdown of P2X7 receptor) were screened, and their tumorigenicity was detected in vivo. The findings of this study demonstrate that both LLC and LA795 cells expressed functional P2X7 receptors, and overexpression of P2X7 receptors promoted the migration and invasion of LLC and LA795 cells. Conversely, the knockdown of the P2X7 receptor yielded contrasting effects. The mechanism involved phosphatidylinositol 3-kinase/protein kinase B/glycogen synthase kinase 3 beta (PI3K/Akt/GSK-3β), c-Jun N-terminal kinase (JNK) signaling pathway and epithelial-mesenchymal transition (EMT). In addition, the knockdown of the P2X7 receptor suppressed cell proliferation and promoted apoptosis in both cells (LLC and LA795). In vivo experiments corroborated these findings, demonstrating that overexpression of the P2X7 receptor promoted tumor growth while its knockdown inhibited tumor growth. The expression levels of related signaling proteins (PI3K/Akt/GSK-3β, JNK, and EMT) in vivo were consistent with the trends observed in vitro. In conclusion, our results suggest that downregulating P2X7 receptor expression can effectively suppress tumor growth, invasion, and migration in NSCLC. Our results suggest that the P2X7 receptor has the potential as a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Qingqing Yu
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Xiaoxiang Peng
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Geng Xu
- Department of Thoracic Surgery, Heze Municipal Hospital, Heze, 274031, Shandong, China.
| | - Xue Bai
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China; Laboratory Department, WeiFang Mental Health Center, Weifang, 261072, Shandong, China.
| | - Yahui Cao
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Yanan Du
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Xin Wang
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Ronglan Zhao
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
2
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025:10.1038/s41568-025-00803-0. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
3
|
Ye F, Xu Y, Zhu X, Ding Q, Wang Y, Lu S, Chen Y. The mechanism of E3 ubiquitin ligase HERC1 regulating ferroptosis in lung adenocarcinoma cells. Cancer Genet 2025; 292-293:92-99. [PMID: 39983667 DOI: 10.1016/j.cancergen.2025.02.001] [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: 07/08/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 02/23/2025]
Abstract
OBJECTIVE Lung adenocarcinoma (LUAD) is the most prevalent subtype of lung cancer. Herein, we probed into the role of E3 ubiquitin protein ligase family member 1 (HERC1) in promoting ferroptosis and inhibiting LUAD cell proliferation by regulating RAF proto-oncogene serine/threonine-protein kinase (C-RAF). METHODS In cultured human normal lung epithelial cells and non-small cell lung adenocarcinoma cell lines, HERC1 expression was determined by RT-qPCR and Western blot tests. PC-9 and Calu-3 cells were transfected with oe-HERC1, oe-C-RAF or their negative controls. Reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH), and Fe2+ levels were assessed by biochemical assays. Cell viability, death, and proliferation were evaluated by CCK-8, LDH and colony formation assays, followed by assessments of HERC1-C-RAF interaction, C-RAF ubiquitin level, and C-RAF protein stability. RESULTS HERC1 was poorly expressed in LUAD cells. HERC1 promoted LUAD cell ferroptosis and repressed their proliferation and migration, corresponding to reduced levels of system xc-, GPX4, and GSH, as well as elevated levels of ROS, MDA, Fe2+, and ACSL4. LUAD cells overexpressing HERC1 displayed decreased C-RAF protein level, HERC1-C-RAF interaction, elevated C-RAF ubiquitin level, and accelerated C-RAF protein degradation, indicating that HERC1 facilitated C-RAF ubiquitin degradation and attenuated C-RAF protein stability via interaction with C-RAF. C-RAF overexpression partially abrogated the regulatory impact of HERC1 on LUAD cell ferroptosis and proliferation. CONCLUSION HERC1 expedites C-RAF ubiquitin degradation by interacting with C-RAF, which consequently promotes ferroptosis, thereby inhibiting LUAD cell proliferation.
Collapse
Affiliation(s)
- Fei Ye
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yi Xu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xujuan Zhu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Qifeng Ding
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yifei Wang
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Songhua Lu
- Haian People's Hospital Department of Thoracic Surgery, Nantong, 226000, Jiangsu, China
| | - Yongbing Chen
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
4
|
Zhao QY, Liu WJ, Wang JG, Li H, Lv JL, Wang Y, Wang C. Increasing cisplatin exposure promotes small-cell lung cancer transformation after a shift from glucose metabolism to fatty acid metabolism. J Cancer Res Clin Oncol 2025; 151:126. [PMID: 40155472 PMCID: PMC11953189 DOI: 10.1007/s00432-025-06164-3] [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: 02/07/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVES Lung cancer is a leading cause of global cancer mortality. Clinical observations reveal that histological transformation from non-small cell lung cancer (NSCLC) to small cell lung cancer (SCLC) is accompanied by mutations in TP53 and RB1. By applying gradually increasing cisplatin concentrations to mimic the escalating drug pressure within the tumor microenvironment, this study investigated the link between phenotypic transformation to SCLC in cisplatin-resistant human lung adenocarcinoma cells and alterations in cellular energy production pathways. MATERIALS AND METHODS We established two cisplatin-resistant NSCLC cell lines with varying resistance levels. RNAseq analyses identified TP53 and RB1 gene mutations. Comprehensive functional assays were performed to characterize A549/DDP1 μg/mL and A549/DDP3 μg/mL cells, focusing on proliferation and migratory capabilities. Cellular bioenergetics were assessed through glycolysis and oxidative phosphorylation analyses. Western blotting was employed to examine epithelial-mesenchymal transition (EMT), glucose metabolism, and lipid metabolism markers. Cell cycle distribution was analyzed by flow cytometry. Additionally, a xenograft mouse model was developed for in vivo validation. RESULTS TP53 and RB1 mutations were associated with cisplatin concentration-dependent phenotypic transformation, with A549/DDP cells acquiring a more aggressive SCLC-like phenotype (In the article we call the A549/DDPSCLC cells). Analysis of cell bioenergetics profiling and Western blot analyses revealed enhanced glucose metabolism in A549/DDP1 μg/mL cells, while A549/DDPSCLC cells exhibited predominant lipid metabolism. Compound3K and Etomoxir specifically inhibit the activity of PKM2 and CPT1A, respectively, with Etomoxir demonstrating substantially inhibited A549/DDPSCLC cells growth and more cell cycle arrest in the G0/G1 phase. Combinatorial of Compound3K and Etomoxir effectively induced cell death in A549/DDPSCLC phenotype cells in vitro. Etomoxir alone or combined with Compound3K significantly inhibited tumor growth in vivo, with enhanced efficacy in the combination group. CONCLUSIONS This study provides the first evidence of cisplatin concentration-dependent metabolic reprogramming during NSCLC-to-SCLC transformation. We identified a phenotypic transition from NSCLC to SCLC accompanied by a metabolic shift from glucose to fatty acid metabolism, offering new insights into therapeutic strategies for treatmentresistant lung cancer.
Collapse
MESH Headings
- Cisplatin/pharmacology
- Humans
- Lung Neoplasms/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Animals
- Small Cell Lung Carcinoma/pathology
- Small Cell Lung Carcinoma/metabolism
- Small Cell Lung Carcinoma/drug therapy
- Small Cell Lung Carcinoma/genetics
- Mice
- Glucose/metabolism
- Fatty Acids/metabolism
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Xenograft Model Antitumor Assays
- Antineoplastic Agents/pharmacology
- Drug Resistance, Neoplasm
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
- Cell Proliferation/drug effects
- Mutation
- Mice, Nude
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Cell Line, Tumor
- Epithelial-Mesenchymal Transition/drug effects
- A549 Cells
Collapse
Affiliation(s)
- Qiu-Yu Zhao
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Wen-Jun Liu
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Jian-Guang Wang
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - He Li
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Jia-Lu Lv
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Yumeng Wang
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, 201210, China
| | - Chun Wang
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China.
| |
Collapse
|
5
|
Luo X, Zhang X, Su D, Li H, Zou M, Xiong Y, Yang L. Deep Clustering-Based Metabolic Stratification of Non-Small Cell Lung Cancer Patients Through Integration of Somatic Mutation Profile and Network Propagation Algorithm. Interdiscip Sci 2025:10.1007/s12539-025-00699-2. [PMID: 40100545 DOI: 10.1007/s12539-025-00699-2] [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: 09/30/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/20/2025]
Abstract
As a common malignancy of the lower respiratory tract, non-small cell lung cancer (NSCLC) represents a major oncological challenge globally, characterized by high incidence and mortality rates. Recent research highlights the critical involvement of somatic mutations in the onset and development of NSCLC. Stratification of NSCLC patients based on somatic mutation data could facilitate the identification of patients likely to respond to personalized therapeutic strategies. However, stratification of NSCLC patients using somatic mutation data is challenging due to the sparseness of this data. In this study, based on sparse somatic mutation data from 4581 NSCLC patients from the Memorial Sloan Kettering Cancer Center (MSKCC) database, we systematically evaluate the metabolic pathway activity in NSCLC patients through the application of network propagation algorithm and computational biology algorithms. Based on these metabolic pathways associated with prognosis, as recognized through univariate Cox regression analysis, NSCLC patients are stratified using the deep clustering algorithm to explore the optimal classification strategy, thereby establishing biologically meaningful metabolic subtypes of NSCLC patients. The precise NSCLC metabolic subtypes obtained from the network propagation algorithm and deep clustering algorithm are systematically evaluated and validated for survival benefits of immunotherapy. Our research marks progress towards developing a universal approach for classifying NSCLC patients based solely on somatic mutation profiles, employing deep clustering algorithm. The implementation of our research will help to deepen the analysis of NSCLC patients' metabolic subtypes from the perspective of tumor microenvironment, providing a strong basis for the formulation of more precise personalized treatment plans.
Collapse
Affiliation(s)
- Xu Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xinpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Dongqing Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Honghao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Min Zou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuqiang Xiong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
6
|
Wu X, Zhang J, Yoshida Y. Disentangling the effects of various risk factors and trends in lung cancer mortality. Sci Rep 2025; 15:8719. [PMID: 40082525 PMCID: PMC11906585 DOI: 10.1038/s41598-025-92373-2] [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: 06/09/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
Lung cancer is a leading cause of mortality in oncological classifications, yet the impact of various risk factors on lung cancer mortality (LCM) in non-smokers remains unclear. This study aims to weigh out the diverse impact of multiple risk factors on LCM rates and identify trends in LCM rates worldwide. We initially employed Random Forest Tree (RFT) and Gradient Boosting Regression (GBR) to identify common primary factors influencing LCM. After eliminating four common primary factors, a comparative analysis between partial and Pearson correlations was conducted to filter out significant factors in the correlations between risk factors and LCM rates across 204 countries from 2005 to 2019. The findings show that excluding the impacts of occupational exposure to arsenic, smoking, residential radon, occupational exposure to silica, occupational exposure to asbestos, high systolic blood pressure, secondhand smoke, child wasting, and alcohol use had a considerably greater impact on LCM than particular matter pollution (PM2.5). Furthermore, a Multiple Joinpoint Regression analysis identified increasing trends of LCM rates in the 142 countries (e.g., China and India); decreasing trends in 38 countries (e.g., Denmark and Norway), and stable trends in 24 countries (e.g., Sudan, Mali, and Australia). This research suggests that in addition to considering the effects of occupational exposure to arsenic, smoking, residential radon, and occupational exposure to silica on LCM rates, occupational exposure to asbestos, high systolic blood pressure, secondhand smoke, child wasting, and alcohol use should be considered in lung cancer prevention strategies, especially in countries with increasing trends of LCM rates.
Collapse
Affiliation(s)
- Xiu Wu
- School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| | - Jinting Zhang
- School of Resource and Environmental Science, Wuhan University, Wuhan, 430070, China.
| | - Yilin Yoshida
- School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
7
|
Jiang H, Zhu T, Chang Z, Liu Z, Ou W, Wang S. A Recurrent Small Cell Lung Carcinoma Harboring an EML4-ALK Fusion Mutation with Sustained Response to Ensartinib: A Case Report. Curr Oncol 2025; 32:163. [PMID: 40136367 PMCID: PMC11941435 DOI: 10.3390/curroncol32030163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine tumor. Lung cancer patients with ALK and EML4 fusions respond significantly to ALK inhibitors. The EML4-ALK fusion gene mutation is the result of an inversion of chromosome 2, which juxtaposes the 5 end of the EML4 gene with the 3 end of the ALK gene. In SCLC, the frequency of fusion genes is very low, and to the best of our knowledge, only four cases of ALK fusion gene mutations in SCLC have been reported. In this report, we describe the treatment of a 74-year-old female patient with SCLC who developed recurrence of hilar lymph node metastasis three years after surgical resection. Postoperative NGS showed that this patient is a SCLC patient harboring a rare EML4-ALK fusion mutation, and a satisfactory 43-month overall survival (OS) was achieved after treatment with ensartinib targeting the EML4-ALK fusion gene mutation. The ALK-TKI may be a new treatment option for these patients. This article provides a therapeutic reference.
Collapse
Affiliation(s)
| | | | | | | | | | - Siyu Wang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Yuexiu District, Guangzhou 510060, China; (H.J.); (T.Z.); (Z.C.); (Z.L.); (W.O.)
| |
Collapse
|
8
|
Lu Y, Fang D, Guo J, Huang H. Partial transformation from non-small cell lung cancer to small cell lung cancer: a case report and literatures review. Front Oncol 2025; 15:1441182. [PMID: 40134592 PMCID: PMC11933710 DOI: 10.3389/fonc.2025.1441182] [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: 05/30/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
A fraction of lung adenocarcinoma patients with gene mutations who receive targeted therapy would experience acquired resistance and undergo small cell lung cancer (SCLC) transformation. The mechanisms behind the transformation of tumor pathological types and the treatment strategies are not fully clear. There have been case reports of the transformation from adenocarcinoma to SCLC, but the partial transformation from adenocarcinoma to SCLC has not been reported. We reported a case of a patient with partial transformation from lung adenocarcinoma to SCLC for the first time. The patient was diagnosed as lung adenocarcinoma with epidermal growth factor receptor (EGFR) 19 exon mutation and Tumor protein p53 (TP53) mutation. She received Epithelial growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) treatment. However, the tumor progression occurred and the lung aspiration pathology revealed a transformation from non-small cell lung cancer (NSCLC) to SCLC. The treatment regimen was changed to cisplatin and etoposide (EP) chemotherapy, resulting in a 2-month PFS. It was worth mentioning that adenocarcinoma cells were found in the patient's emerging pericardial effusion, suggesting the co-existence of both adenocarcinoma and SCLC components. This is the first report of partial transformation from NSCLC to SCLC in the context of definitive pathology. It highlights that when no more pathological biopsy is feasible, we should be alert to the partial transformation and adopt the appropriate treatment.
Collapse
Affiliation(s)
| | | | | | - Huaqiong Huang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Kuang L, Wang P, Zhou L, Li Y. Transformation of lung adenocarcinoma to small cell lung cancer following osimertinib treatment: a case report and literature review. Anticancer Drugs 2025; 36:253-259. [PMID: 39792045 PMCID: PMC11781543 DOI: 10.1097/cad.0000000000001686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/09/2024] [Indexed: 01/12/2025]
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) effectively treat EGFR-mutant lung adenocarcinoma, demonstrating initial efficacy but eventually leading to acquired resistance. Small cell transformation is a rare resistance mechanism to EGFR-TKIs in lung adenocarcinoma, which can complicate clinical diagnosis and treatment. We present a patient with lung adenocarcinoma who underwent a prior pneumonectomy and adjuvant chemotherapy and was treated with osimertinib after the recurrence of lung cancer. Small cell transformation occurred approximately 20 months after starting osimertinib treatment. After this transformation, the patient underwent lung radiotherapy and cisplatin-etoposide chemotherapy, which stabilized the disease. Following the confirmation of small cell lung cancer (SCLC) via thyroid puncture, treatments with irinotecan, irinotecan plus atezolizumab, thyroid radiotherapy, adrenal radiotherapy, and head radiotherapy were sequentially administered, yet the disease continued to progress. The patient succumbed to the disease in May 2023 because of progression and organ failure, with an overall survival of 52.7 months, including 16 months post small cell transformation. This case highlights the possibility of osimertinib causing lung adenocarcinoma to transform into SCLC and underscores rebiopsies' importance in identifying resistance mechanisms to EGFR-TKIs. Increased levels of neuron-specific enolase and pro-gastrin releasing peptide can signal early transformation into SCLC.
Collapse
Affiliation(s)
- Linwu Kuang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhou
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangkai Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Al Bakir M, Reading JL, Gamble S, Rosenthal R, Uddin I, Rowan A, Przewrocka J, Rogers A, Wong YNS, Bentzen AK, Veeriah S, Ward S, Garnett AT, Kalavakur P, Martínez-Ruiz C, Puttick C, Huebner A, Cook DE, Moore DA, Abbosh C, Hiley CT, Naceur-Lombardelli C, Watkins TBK, Petkovic M, Schwarz RF, Gálvez-Cancino F, Litchfield K, Meldgaard P, Sorensen BS, Madsen LB, Jäger D, Forster MD, Arkenau T, Domingo-Vila C, Tree TIM, Kadivar M, Hadrup SR, Chain B, Quezada SA, McGranahan N, Swanton C. Clonal driver neoantigen loss under EGFR TKI and immune selection pressures. Nature 2025; 639:1052-1059. [PMID: 39972134 PMCID: PMC11946900 DOI: 10.1038/s41586-025-08586-y] [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: 12/16/2022] [Accepted: 01/02/2025] [Indexed: 02/21/2025]
Abstract
Neoantigen vaccines are under investigation for various cancers, including epidermal growth factor receptor (EGFR)-driven lung cancers1,2. We tracked the phylogenetic history of an EGFR mutant lung cancer treated with erlotinib, osimertinib, radiotherapy and a personalized neopeptide vaccine (NPV) targeting ten somatic mutations, including EGFR exon 19 deletion (ex19del). The ex19del mutation was clonal, but is likely to have appeared after a whole-genome doubling (WGD) event. Following osimertinib and NPV treatment, loss of the ex19del mutation was identified in a progressing small-cell-transformed liver metastasis. Circulating tumour DNA analyses tracking 467 somatic variants revealed the presence of this EGFR wild-type clone before vaccination and its expansion during osimertinib/NPV therapy. Despite systemic T cell reactivity to the vaccine-targeted ex19del neoantigen, the NPV failed to halt disease progression. The liver metastasis lost vaccine-targeted neoantigens through chromosomal instability and exhibited a hostile microenvironment, characterized by limited immune infiltration, low CXCL9 and elevated M2 macrophage levels. Neoantigens arising post-WGD were more likely to be absent in the progressing liver metastasis than those occurring pre-WGD, suggesting that prioritizing pre-WGD neoantigens may improve vaccine design. Data from the TRACERx 421 cohort3 provide evidence that pre-WGD mutations better represent clonal variants, and owing to their presence at multiple copy numbers, are less likely to be lost in metastatic transition. These data highlight the power of phylogenetic disease tracking and functional T cell profiling to understand mechanisms of immune escape during combination therapies.
Collapse
Affiliation(s)
- Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James L Reading
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Samuel Gamble
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Rachel Rosenthal
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Imran Uddin
- Division of Infection and Immunity, University College London, London, UK
| | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Joanna Przewrocka
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Amber Rogers
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Yien Ning Sophia Wong
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Amalie K Bentzen
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | | | | | - Carlos Martínez-Ruiz
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| | - Clare Puttick
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| | - Ariana Huebner
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| | - Daniel E Cook
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - David A Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospital NHS Foundation Trust, London, UK
| | - Chris Abbosh
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Crispin T Hiley
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Marina Petkovic
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Biology, Humboldt University of Berlin, Berlin, Germany
- Division of Oncology and Hematology, Department of Pediatrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Roland F Schwarz
- Institute for Computational Cancer Biology (ICCB), Center for Integrated Oncology (CIO), Cancer Research Center Cologne Essen (CCCE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
| | - Felipe Gálvez-Cancino
- Immune-Regulation and Immune-Interactions Laboratory, Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Headington, UK
| | - Kevin Litchfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Line Bille Madsen
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin D Forster
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Oncology, UCL Cancer Institute, London, UK
| | | | - Clara Domingo-Vila
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Timothy I M Tree
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mohammad Kadivar
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Benny Chain
- Division of Infection and Immunity, University College London, London, UK
- Department of Computer Sciences, University College London, London, UK
| | - Sergio A Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK.
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK.
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
11
|
Jiang Z, Sun X, Li Y, Wang J, Wang C, Pan Z, Yang Y. Anlotinib induced ferroptosis through the p53/xCT/GPX4 pathway in non-small cell lung cancer. Transl Oncol 2025; 53:102289. [PMID: 39827731 PMCID: PMC11787696 DOI: 10.1016/j.tranon.2025.102289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
Anlotinib, an anti-angiogenic agent, has demonstrated significant anti-tumor effects in non-small cell lung cancer (NSCLC). However, whether anlotinib exerts its anti-tumor activity in NSCLC through ferroptosis, and its underlying mechanisms, remain unclear. This study revealed that anlotinib effectively inhibited the proliferation of NSCLC cells in a time- and dose-dependent manner. Treatment with anlotinib resulted in increased levels of ferroptosis targets (lipid reactive oxygen species and malondialdehyde) and p53 protein expression, while decreasing glutathione levels and the protein expression of solute carrier family 7 member 11 (xCT) and glutathione peroxidase 4 (GPX4). Notably, the ferroptosis inhibitor, Ferrostatin-1 (Fer-1), or the p53 inhibitor, Pifithrin-α (PFT-α), reversed the observed effects on ferroptosis induction in NSCLC cells. Consistently, our in vivo studies showed accelerated tumor growth rates for the anlotinib/Fer-1 group and the anlotinib/PFT-α group compared with administration of anlotinib alone. However, anlotinib-induced ferroptosis was suppressed in p53-deficient cells. Collectively, these findings confirm that anlotinib exerts potent anti-tumor effects both in vitro and in vivo by inducing ferroptosis by modulating the p53/xCT/GPX4 pathway specifically within NSCLC cells.
Collapse
Affiliation(s)
- Zhansheng Jiang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin China.
| | - Xinge Sun
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin China
| | - Yuan Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiahe Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Cong Wang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin China
| | - Zhanyu Pan
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin China
| | - Yinli Yang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin China.
| |
Collapse
|
12
|
Richlitzki C, Manapov F, Holzgreve A, Rabe M, Werner RA, Belka C, Unterrainer M, Eze C. Advances of PET/CT in Target Delineation of Lung Cancer Before Radiation Therapy. Semin Nucl Med 2025; 55:190-201. [PMID: 40064578 DOI: 10.1053/j.semnuclmed.2025.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
In the clinical management of lung cancer, radiotherapy remains a cornerstone of multimodal treatment strategies, often used alongside surgery or in combination with systemic therapies such as chemotherapy, tyrosine kinase inhibitors, and immune checkpoint inhibitors. While conventional imaging modalities like computed tomography (CT) and magnetic resonance imaging (MRI) continue to play a central role in staging, response assessment, and radiotherapy planning, advanced imaging techniques, particularly [18F]FDG PET/CT, are being increasingly integrated into routine clinical practice. These advanced techniques address the limitations of standard imaging by providing insight into molecular and metabolic tumor characteristics, enabling precise tumor visualization, accurate target volume delineation, and early treatment response assessment. This review examines the role of radiotherapy in the multidisciplinary management of lung cancer, detailing current concepts of morphological and functional imaging for staging and treatment planning. It also highlights the growing importance of PET-based radiotherapy planning, emphasizing its contributions to target volume definition and predictive value for treatment outcomes. Recent methodological advances, including the integration of artificial intelligence (AI), radiomics, technical innovations, and novel PET ligands, are discussed, highlighting their potential to improve the precision, efficacy, and personalization of lung cancer radiotherapy planning.
Collapse
Affiliation(s)
- Cedric Richlitzki
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Ahmanson Translational Theranostics Division, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Moritz Rabe
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rudolf Alexander Werner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany; Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany; Bavarian Cancer Research Center, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Die Radiologie, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
13
|
Diao S, Wan Y, Huang D, Huang S, Sadiq T, Khan MS, Hussain L, Alkahtani BS, Mazhar T. Optimizing Bi-LSTM networks for improved lung cancer detection accuracy. PLoS One 2025; 20:e0316136. [PMID: 39992919 PMCID: PMC11849851 DOI: 10.1371/journal.pone.0316136] [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: 09/30/2024] [Accepted: 12/05/2024] [Indexed: 02/26/2025] Open
Abstract
Lung cancer remains a leading cause of cancer-related deaths worldwide, with low survival rates often attributed to late-stage diagnosis. To address this critical health challenge, researchers have developed computer-aided diagnosis (CAD) systems that rely on feature extraction from medical images. However, accurately identifying the most informative image features for lung cancer detection remains a significant challenge. This study aimed to compare the effectiveness of both hand-crafted and deep learning-based approaches for lung cancer diagnosis. We employed traditional hand-crafted features, such as Gray Level Co-occurrence Matrix (GLCM) features, in conjunction with traditional machine learning algorithms. To explore the potential of deep learning, we also optimized and implemented a Bidirectional Long Short-Term Memory (Bi-LSTM) network for lung cancer detection. The results revealed that the highest performance using hand-crafted features was achieved by extracting GLCM features and utilizing Support Vector Machine (SVM) with different kernels, reaching an accuracy of 99.78% and an AUC of 0.999. However, the deep learning Bi-LSTM network surpassed both methods, achieving an accuracy of 99.89% and an AUC of 1.0000. These findings suggest that the proposed methodology, combining hand-crafted features and deep learning, holds significant promise for enhancing early lung cancer detection and ultimately improving diagnosis systems.
Collapse
Affiliation(s)
- Su Diao
- Department of Industrial & Systems Engineering, Auburn University, Auburn, Alabama, United States of America
| | - Yajie Wan
- Department of Computer Science, Brown University, Providence, RI, United States of America
| | - Danyi Huang
- Department of Chemical Engineering, Columbia University, New York City, NY, United States of America
| | - Shijia Huang
- Fu Foundation School of Engineering and Applied Science, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, United States of America
| | - Touseef Sadiq
- Department of Information and Communication Technology, Centre for Artificial Intelligence Research (CAIR), University of Agder, Grimstad, Norway
| | | | - Lal Hussain
- Department of Computer Science and Information Technology, The University of Azad Jammu and Kashmir, Chattar Kalas Campus, Muzaffarabad, Pakistan
- Department of Computer Science, Neelum Campus, The University of Azad Jammu and Kashmir, Azad Kashmir, Pakistan
| | - Badr S. Alkahtani
- Department of Mathematics, King Saud University, Riyadh, Saudi Arabia
| | - Tehseen Mazhar
- School of Computer Science, National College of Business Administration and Economics, Lahore, Pakistan
- Department of Computer Science and Information Technology, School Education Department, Government of Punjab, Layyah, Pakistan
| |
Collapse
|
14
|
Nambirajan A, Rathor A, Baskarane H, Gs A, Khurana S, Sushmitha S, Sharma A, Malik PS, Jain D. Baseline retinoblastoma transcriptional corepressor 1 (Rb1) functional inactivation is a pre-requisite but not sufficient for small-cell histological transformation in epidermal growth factor receptor (EGFR) mutant lung adenocarcinomas post-tyrosine kinase inhibitor therapy. Virchows Arch 2025:10.1007/s00428-025-04054-0. [PMID: 39982521 DOI: 10.1007/s00428-025-04054-0] [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: 01/29/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Small-cell transformation is an uncommon mechanism of tyrosine receptor kinase inhibitor (TKI) resistance in epidermal growth factor receptor (EGFR)-mutant lung adenocarcinomas. This study aims to assess the dynamic changes in the molecular landscape and Rb1 functional status in EGFR-mutant lung adenocarcinomas transforming to small-cell carcinomas post-treatment with EGFR-TKIs. This is an ambispective study (2019-2023) wherein the baseline and post-TKI biopsies of EGFR-mutant lung adenocarcinomas with small-cell transformation were subject to Rb1 immunohistochemistry and 72-gene targeted panel next-generation sequencing. Rb1-deficiency was defined as Rb1 protein loss or Rb1retained/p16high/Cyclin-D1low protein expression profile with RB1 mutations. A cohort of EGFR-mutant lung adenocarcinomas without small-cell transformation was included for Rb1 status comparison. Small-cell transformation was diagnosed in 9 patients (10%, 9/84) on their post-TKI biopsy. All their tested baseline adenocarcinoma (n = 7) and post-TKI small-cell carcinoma (n = 9) samples were Rb1-deficient, with additional TP53 (11/11) and PTEN mutations (2/11). Eighteen paired samples from 9 patients without small-cell transformation revealed Rb1-deficiency in one patient (1/9) only. Baseline functional inactivation of Rb1 is nearly universal in EGFR-mutant adenocarcinomas transforming to small-cell carcinomas post-EGFR TKI suggesting that Rb1 loss is prerequisite for small-cell transformation. However, it is likely not sufficient as not all adenocarcinomas with baseline Rb1 loss transform into small-cell carcinomas. Except for TP53 and PTEN, recurrent mutations in other common oncogenes tested were not detected at baseline or at progression. Within the limitation of a small sample size, specific molecular events that drive small-cell transformation remain unclear.
Collapse
Affiliation(s)
- Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Amber Rathor
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Hemavathi Baskarane
- Department of Medical Oncology, IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Anju Gs
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Sachin Khurana
- Department of Medical Oncology, IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Somagattu Sushmitha
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Aparna Sharma
- Department of Medical Oncology, National Cancer Institute, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhat Singh Malik
- Department of Medical Oncology, IRCH, All India Institute of Medical Sciences, New Delhi, India.
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
15
|
Cheng K, Zhu Y, Sang R, Kuang Z, Cao Y. Case report: A patient with EGFR L861Q positive adenosquamous lung carcinoma transforming into large cell neuroendocrine cancer after treatment with Almonertinib. Front Oncol 2025; 15:1453066. [PMID: 40040728 PMCID: PMC11876032 DOI: 10.3389/fonc.2025.1453066] [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: 06/22/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Almonertinib, a third-generation epidermal growth factor receptor tyrosine kinase inhibitor, is selective for both epidermal growth factor receptor tyrosine kinase inhibitor-sensitizing and T790M resistance mutations. However, resistance to the third-generation EGFR-TKIs is still inevitable. Econdary EGFR mutations, and bypass pathway activation have been reported with Almonertinib therapy. This article presents a rare case report of a patient with EGFR L861Q positive adenosquamous lung carcinoma who transformed into large cell neuroendocrine carcinoma following treatment with Almonertinib. The patient exhibited disease progression 8 months after initiating Almonertinib treatment, and a blood genetic test revealed mutations in EGFR L861Q and EGFR L858R. A subsequent lung biopsy after progression confirmed the diagnosis of large cell neuroendocrine carcinoma, and subsequently treatment with cisplatin and etoposide was effective. Transformation into neuroendocrine carcinoma is one of the mechanisms behind resistance to Almonertinib in adenosquamous lung carcinoma. EGFR mutations may persist even after transformation into neuroendocrine carcinoma. For non-small cell lung cancer patients undergoing Almonertinib therapy, this case report emphasizes the importance of performing a timely pathological biopsy upon the emergence of resistance.
Collapse
Affiliation(s)
- Kele Cheng
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Zhu
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ran Sang
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongsheng Kuang
- Department of Oncology Center, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Cao
- Department of Oncology Center, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Wang J, Li X, Dong S, Hu S, Ran F, Qian Y. Case report: Personalized management of treatment resistance in advanced NSCLC patients with mutated epidermal growth factor receptor: special examples and literature review. Front Oncol 2025; 15:1525881. [PMID: 39995840 PMCID: PMC11847684 DOI: 10.3389/fonc.2025.1525881] [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: 11/10/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
The treatment landscape of non-small cell lung cancer (NSCLC) has shifted significantly from empirical, histology-driven, and clinician-directed cytotoxic regimens to a stratified approach predicated on molecular profiling of tumor genetics and immune biomarkers, by the former can indicate targeted therapy that bull's eye hits the arrow, while the latter can hint the benefit amplitude of immune checkpoint inhibitors (ICBs). While third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) have become the cornerstone of frontline therapy for patients harboring classic sensitive EGFR mutations, all tumors ultimately develop acquired resistance to these approaches, which can be categorized into three primary mechanism subclasses. The first subclass involves the acquisition of target mutations that lead to changes in the kinase domain, thereby hindering drug binding. The second mechanism, known as bypass resistance, entails tumor clones utilizing alternative signaling pathways for proliferation. Lastly, the third acquired mechanism pertains to histological transformation, such as the emergence of small cell lung cancer (SCLC) clones. The transformation of pathological types has brought great confusion to the clinical diagnosis and treatment process. We report a case of advanced lung adenocarcinoma with EGFR-sensitive mutation that transformed into small cell lung cancer after EGFR-TKIs treatment. Subsequent treatment revealed the presence of both adenocarcinoma and small cell carcinoma through needle biopsies at various metastatic sites. Based on the pathological, the patient received combination therapy with anlotinib at different times and achieved a long survival time.
Collapse
Affiliation(s)
- Jun Wang
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Natural Medicine and Molecular Engineering, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaojing Li
- Department of Pharmacy, Puren Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Shuang Dong
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengming Ran
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Qian
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Li M, Wang X, Gong J, Lu H. The analysis of molecular classification of pulmonary neuroendocrine tumors and relationship between YAP1 and efficacy. Invest New Drugs 2025; 43:108-117. [PMID: 39786663 DOI: 10.1007/s10637-024-01492-6] [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: 10/12/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
A novel molecular classification for small cell lung cancer (SCLC) has been established utilizing the transcription factors achaete-scute homologue 1 (ASCL1), neurogenic differentiation factor 1 (NeuroD1), POU class 2 homeobox 3 (POU2F3), and yes-associated protein 1 (YAP1). This classification was predicated on the transcription factors. Conversely, there is a paucity of information regarding the distribution of these markers in other subtypes of pulmonary neuroendocrine tumors (PNET). Clinical and survival data for PNET patients were gathered from January 2008 to December 2020. Immunohistochemical analysis was employed to evaluate the expression. The relationship between YAP1 expression and outcomes in patients with pulmonary large cell neuroendocrine carcinoma (LCNEC) was examined. Data from low-grade PNET patients who had previously undergone immunotherapy were retrospectively gathered and analyzed. The ASCL1 positive rate was markedly elevated in SCLC (7.1% vs. 60%; P < 0.001) and LCNEC patients (7.1% vs. 38.5%; P = 0.034) compared to PC patients. The YAP1-positive rate was elevated in LCNEC compared to SCLC (43.6% vs. 20%, P = 0.028) and pulmonary carcinoid (PC) patients (43.6% vs. 21.4%; P = 0.021). The DLL3-positive rate in SCLC patients was greater than in SCLC and PC patients (37.1% vs. 23.1% vs. 0%; P = 0.028, P = 0.021). A significant level of tumor heterogeneity was noted, with SCLC and LCNEC patients exhibiting markedly higher heterogeneity than PC patients (65.7% vs. 56.3% vs. 21.4%; P = 0.005, P = 0.025). In patients with LCNEC, YAP1 positivity exhibited no correlation with PD-L1 expression (17.1% vs. 45.7%, P = 0.518). Tumor heterogeneity was also noted in transformed SCLC, with no significant differences in the expression levels of transcription factors between transformed and traditional SCLC. In 13 LCNEC patients with a history of ICI application, YAP1 exhibited no significant effect on PFS (P = 0.331) or OS (P = 0.17) in the subgroup analysis of LCNEC patients. Among the 14 patients with low-grade PNET who underwent immunotherapy, the disease control rate was 85.7%. Patients with high-grade PNET have high levels of expression of ASCL1 and DLL3, whereas patients with LCNEC have high levels of expression of YAP1. With regard to the transcription factor level, it was found that patients with SCLC and LCNEC had a much higher degree of tumor heterogeneity than those with PC. In patients with LCNEC who were receiving monotherapy of ICIs or chemotherapy in combination with ICIs, the expression of YAP1 did not appear to have any clear impact on the prognosis. This is due to the limited sample size of the study, which requires additional investigation. When compared to the expression of TFs in regular SCLC, the expression of TFs in converted SCLC is comparable.
Collapse
Affiliation(s)
- Meihui Li
- Department of Radiotherapy, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Xinyuan Wang
- Postgraduate Training Base Alliance, Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Jiali Gong
- Department of Hematology and Oncology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Hongyang Lu
- Postgraduate Training Base Alliance, Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China.
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
18
|
Lasvergnas J, Monnet I, Auliac JB, Rousseau-Bussac G, Chouaid C, Assié JB. Recurrence as a small cell lung cancer transformation in a resected stage IIIA EGFR-mutated non-small cell lung cancer treated with adjuvant osimertinib: a case report. Transl Lung Cancer Res 2025; 14:287-291. [PMID: 39958211 PMCID: PMC11826274 DOI: 10.21037/tlcr-24-830] [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: 09/12/2024] [Accepted: 11/26/2024] [Indexed: 02/18/2025]
Abstract
Background Based on improvements in recurrence-free and overall survival, osimertinib is now widely used as an adjuvant treatment in stage II-IIIA non-small cell lung cancer (NSCLC) presenting with a common epithelial growth factor receptor (EGFR) mutation. Histological transformation is a well-known resistance mechanism to osimertinib in EGFR-mutated metastatic NSCLC, but we currently have insufficient data on recurrence mechanisms in the adjuvant context. We present here the case of a patient treated with adjuvant osimertinib and presenting a small cell lung cancer (SCLC) transformation as a recurrence. Case Description A 54-year-old man, never-smoker and with no previous medical history, underwent right superior lobectomy with lymph node resection for a pT3N1M0 [stage IIIA, tumor-node-metastasis (TNM) 8th edition] adenocarcinoma. Programmed death-ligand 1 (PD-L1) negative with an EGFR exon 19 deletion. The patient received 4 cycles of adjuvant chemotherapy before starting adjuvant osimertinib 80 mg. After 35 months of adjuvant osimertinib the patient had a local recurrence and the re biopsy showed an SCLC transformation, underlining the importance of careful surveillance and biopsy at the time of recurrence in EGFR-mutated NSCLC. Conclusions this case report provides evidence of SCLC transformation while on adjuvant osimertinib, in a pT3N1 EGFR, RB1 and TP53-mutated pulmonary adenocarcinoma. This highlights the importance of biopsy on recurrence and the transformation potential of the EGFR, RB1 and TP53-mutated adenocarcinomas.
Collapse
Affiliation(s)
- Julie Lasvergnas
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Isabelle Monnet
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jean-Bernard Auliac
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | | | - Christos Chouaid
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jean-Baptiste Assié
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| |
Collapse
|
19
|
Zheng T, Wang S, Liu W, Lu Y. Targeting KAT6A/B as a New Therapeutic Strategy for Cancer Therapy. J Med Chem 2025; 68:1002-1020. [PMID: 39761381 DOI: 10.1021/acs.jmedchem.4c02613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The lysine acetyltransferase 6A (KAT6A, MOZ, MYST3) is a member of the MYST family of protein acetyltransferases, which are essential for different biological processes such as craniofacial, embryonic, stem cell development, and hematopoiesis. KAT6A is an oncogene in human acute myeloid leukemia (AML), and KAT6A overexpression in AML is associated with metastases and poor prognoses. Furthermore, KAT6A mutations play an important role in cancer formation and progression and result in therapeutic resistance in both hematopoietic malignancies and solid tumors. In this review, we discuss the structural and biological functions of KAT6A and summarize the influence of KAT6A in the development of various tumors. In addition, the recent KAT6A/B inhibitors, their anticancer activities, challenges, and perspective of developing KAT6A/B inhibitors as anticancer drug candidates were introduced.
Collapse
Affiliation(s)
- Tianpeng Zheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shanglong Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
20
|
Zhang Y, Li KJ, Wang C, Zou CL, Su M. Brigatinib treatment in a patient with advanced NSCLC with XPO1-ALK fusion: a case report. Front Oncol 2025; 14:1503262. [PMID: 39911820 PMCID: PMC11794119 DOI: 10.3389/fonc.2024.1503262] [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: 09/28/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
Patients with ALK-rearranged non-small cell lung cancer (NSCLC) who are treated with ALK tyrosine kinase inhibitors (ALK TKIs) have better prognoses. In this case report, we provide evidence of a novel ALK fusion, XPO1-ALK (intergenic), identified by next-generation DNA sequencing in a patient with advanced lung cancer. After 5 months of brigatinib targeted therapy, the patient clearly experienced tumor disintegration, and this treatment resulted in partial remission. To date, this patient has experienced 5 months of progression-free survival after brigatinib treatment. In addition to reporting the identification of a novel ALK fusion, XPO1-ALK (intergenic), and the sensitivity and safety of brigatinib treatment for lung cancer, this study increased the list of known ALK fusion partners in ALK-positive NSCLC. This case report has a significant clinical reference.
Collapse
Affiliation(s)
| | | | | | | | - Meng Su
- Department of Radiotherapy, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| |
Collapse
|
21
|
Lin F, Luo H, Wang J, Li Q, Zha L. Macrophage-derived extracellular vesicles as new players in chronic non-communicable diseases. Front Immunol 2025; 15:1479330. [PMID: 39896803 PMCID: PMC11782043 DOI: 10.3389/fimmu.2024.1479330] [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: 08/13/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Macrophages are innate immune cells present in all tissues and play an important role in almost all aspects of the biology of living organisms. Extracellular vesicles (EVs) are released by cells and transport their contents (micro RNAs, mRNA, proteins, and long noncoding RNAs) to nearby or distant cells for cell-to-cell communication. Numerous studies have shown that macrophage-derived extracellular vesicles (M-EVs) and their contents play an important role in a variety of diseases and show great potential as biomarkers, therapeutics, and drug delivery vehicles for diseases. This article reviews the biological functions and mechanisms of M-EVs and their contents in chronic non-communicable diseases such as cardiovascular diseases, metabolic diseases, cancer, inflammatory diseases and bone-related diseases. In addition, the potential application of M-EVs as drug delivery systems for various diseases have been summarized.
Collapse
Affiliation(s)
- Fengjuan Lin
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Huiyu Luo
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiexian Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Qing Li
- Department of Clinical Nutrition, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Longying Zha
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Joshi A, Bhaskar N, Pearson JD. Neuroendocrine Transformation as a Mechanism of Resistance to Targeted Lung Cancer Therapies: Emerging Mechanisms and Their Therapeutic Implications. Cancers (Basel) 2025; 17:260. [PMID: 39858043 PMCID: PMC11763869 DOI: 10.3390/cancers17020260] [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: 11/15/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, highlighting a major clinical challenge. Lung cancer is broadly classified into two histologically distinct subtypes, termed small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC). Identification of various oncogenic drivers of NSCLC has facilitated the development of targeted therapies that have dramatically improved patient outcomes. However, acquired resistance to these targeted therapies is common, which ultimately results in patient relapse. Several on-target and off-target resistance mechanisms have been described for targeted therapies in NSCLC. One common off-target mechanism of resistance to these therapies is histological transformation of the initial NSCLC into SCLC, a highly aggressive form of lung cancer that exhibits neuroendocrine histology. This mechanism of resistance presents a significant clinical challenge, since there are very few treatments available for these relapsed patients. Although the phenomenon of NSCLC-to-SCLC transformation was described almost 20 years ago, only recently have we begun to understand the mechanisms underlying this therapy-driven response. These recent discoveries will be key to identifying novel biomarkers and therapeutic strategies to improve outcomes of patients that undergo NSCLC-to-SCLC transformation. Here, we highlight these recent advances and discuss the potential therapeutic strategies that they have uncovered to target this mechanism of resistance.
Collapse
Affiliation(s)
- Asim Joshi
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Nivitha Bhaskar
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Joel D. Pearson
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
23
|
Kranthi Reddy S, Reddy SVG, Hussain Basha S. Discovery of novel PDGFR inhibitors targeting non-small cell lung cancer using a multistep machine learning assisted hybrid virtual screening approach. RSC Adv 2025; 15:851-869. [PMID: 39802474 PMCID: PMC11718652 DOI: 10.1039/d4ra06975g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Non-Small Cell Lung Cancer (NSCLC) is a formidable global health challenge, responsible for the majority of cancer-related deaths worldwide. The Platelet-Derived Growth Factor Receptor (PDGFR) has emerged as a promising therapeutic target in NSCLC, given its crucial involvement in cell growth, proliferation, angiogenesis, and tumor progression. Among PDGFR inhibitors, avapritinib has garnered attention due to its selective activity against mutant forms of PDGFR, particularly PDGFRA D842V and KIT exon 17 D816V, linked to resistance against conventional tyrosine kinase inhibitors. In recent years, Machine Learning has emerged as a powerful tool in pharmaceutical research, offering data-driven insights and accelerating lead identification for drug discovery. In this research article, we focus on the application of Machine Learning, alongside the RDKit toolkit, to identify potential anti-cancer drug candidates targeting PDGFR in NSCLC. Our study demonstrates how smart algorithms efficiently narrow down large screening collections to target-specific sets of just a few hundred small molecules, streamlining the hit discovery process. Employing a Machine Learning-assisted virtual screening strategy, we successfully preselected 220 compounds with potential PDGFRA inhibitory activity from a vast library of 1.048 million compounds, representing a mere 0.013% of the original library. To validate these candidates, we employed traditional genetic algorithm-based virtual screening and docking methods. Remarkably, we found that ZINC000002931631 exhibited comparable or even superior inhibitory potential against PDGFRA compared to Avapritinib, which highlights the value of our Machine Learning approach. Moreover, as part of our lead validation studies, we conducted molecular dynamic simulations, revealing critical molecular-level interactions responsible for the conformational changes in PDGFRA necessary for substrate binding. Our study exemplifies the potential of Machine Learning in the drug discovery process, providing a more efficient and cost-effective means of identifying promising drug candidates for NSCLC treatment. The success of this approach in preselecting compounds with potent PDGFRA inhibitory potential highlights its significance in advancing personalized and targeted therapies for cancer treatment.
Collapse
Affiliation(s)
| | - S V G Reddy
- Department of CSE, GST, GITAM (Deemed to be University) Visakhapatnam A.P India
| | | |
Collapse
|
24
|
Behrouzi R, Clipson A, Simpson KL, Blackhall F, Rothwell DG, Dive C, Mouliere F. Cell-free and extrachromosomal DNA profiling of small cell lung cancer. Trends Mol Med 2025; 31:64-78. [PMID: 39232927 DOI: 10.1016/j.molmed.2024.08.004] [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: 05/30/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024]
Abstract
Small cell lung cancer (SCLC) is highly aggressive with poor prognosis. Despite a relative prevalence of circulating tumour DNA (ctDNA) in SCLC, liquid biopsies are not currently implemented, unlike non-SCLC where cell-free DNA (cfDNA) mutation profiling in the blood has utility for guiding targeted therapies and assessing minimal residual disease. cfDNA methylation profiling is highly sensitive for SCLC detection and holds promise for disease monitoring and molecular subtyping; cfDNA fragmentation profiling has also demonstrated clinical potential. Extrachromosomal DNA (ecDNA), that is often observed in SCLC, promotes tumour heterogeneity and chemotherapy resistance and can be detected in blood. We discuss how these cfDNA profiling modalities can be harnessed to expand the clinical applications of liquid biopsy in SCLC.
Collapse
Affiliation(s)
- Roya Behrouzi
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Alexandra Clipson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Kathryn L Simpson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Dominic G Rothwell
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Florent Mouliere
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
25
|
Yazawa T. What is the cell of origin of lung neuroendocrine carcinoma? Respir Investig 2025; 63:40-41. [PMID: 39632325 DOI: 10.1016/j.resinv.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Takuya Yazawa
- Department of Pathology, Dokkyo Medical University, 880 Kita-kobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, 321-0293, Japan.
| |
Collapse
|
26
|
Zuo Y, Yang P, Yang R, Hou J, Feng R, Liang P, Liu J. Determination of osimertinib concentration in rat plasma and lung/brain tissues. Am J Transl Res 2024; 16:8008-8022. [PMID: 39822485 PMCID: PMC11733393 DOI: 10.62347/syzd2489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/06/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVES The aim of this study was to establish an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method for the detection of osimertinib in rat plasma, lung and brain tissues. METHODS Forty-eight rats were randomly divided into an experimental group (receiving osimertinib at doses of 5, 8, and 10 mg/kg) and a control group. After continuous intragastric administration for 15 days, samples of blood, lung, and brain tissue were collected. Chromatographic separation was achieved using a BEH C18 column with gradient elution, employing a mobile phase of water (containing 0.1% (v/v) formic acid) and acetonitrile. The concentration of osimertinib in the samples was determined using an AB SCIEX 5500 triple quadrupole mass spectrometer operated in positive electrospray ionization (ESI+) and multiple reaction monitoring (MRM) mode. RESULTS A UPLC-MS/MS analytical method for determining osimertinib concentrations was successfully established and validated. A linear relationship was observed for osimertinib concentrations in plasma within the range of 1-300 ng/mL, and in lung and brain tissues within the range of 0.5-50 ng/mL. The selectivity, accuracy, precision, matrix effect, extraction recovery, and stability all meet the requirements of methodological validation criteria. CONCLUSIONS A rapid and sensitive UPLC-MS/MS method was developed and validated for quantifying osimertinib concentrations in rat plasma, lung, and brain tissues, providing a valuable tool for pharmacokinetic and tissue distribution studies.
Collapse
Affiliation(s)
- Yalan Zuo
- Department of Pharmacy, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| | - Peidan Yang
- College of Pharmacy, Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| | - Ruixia Yang
- Department of Pharmacy, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| | - Juan Hou
- Department of Pharmacy, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| | - Rui Feng
- Department of Pharmacy, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| | - Ping Liang
- Department of Pharmacy, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| | - Jiang Liu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, China
| |
Collapse
|
27
|
Boyraz B, Medford A, Ly A, Chebib I. Transdifferentiation of Secretory Carcinoma to Metaplastic Carcinoma at Metastatic Site. Int J Surg Pathol 2024:10668969241300487. [PMID: 39665211 DOI: 10.1177/10668969241300487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Secretory carcinoma is a rare breast tumor driven by ETV6::NTRK3 fusion. It has characteristic morphologic features and identification of these tumors is critical as these patients may benefit from TRK inhibitors. Morphologic shift upon treatment has not been reported in secretory carcinoma or other NTRK-driven tumors. Herein we describe a patient with secretory carcinoma showing transdifferentiation into metaplastic carcinoma (spindle cell and squamous cell carcinoma) at the metastatic site following treatment. Identification of these morphologic shifts is crucial for accurate classification and identification of potential resistant mechanisms.
Collapse
Affiliation(s)
- Baris Boyraz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/New York Presbyterian Hospital, New York, NY, USA
| | - Arielle Medford
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Amy Ly
- James Homer Wright Pathology Laboratories, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivan Chebib
- James Homer Wright Pathology Laboratories, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Ming H, Lu T, Zhou H, Song W, Dai H. Synergistic inhibitory effect of atmospheric pressure plasma and berberine on non‑small cell lung cancer cells via inducing apoptosis. Mol Biol Rep 2024; 52:37. [PMID: 39643828 DOI: 10.1007/s11033-024-10132-4] [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: 10/05/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a type of lung cancer, the incidence and mortality rate have been high, and the use of monotherapy is easy to make patients develop tolerance. Atmospheric pressure plasma (APP) is an emerging technology for killing cancer cells in recent years, and combination of berberine (BBR) mechanism has not been fully elucidated for NSCLC. The article's primary goal is to investigate the effect of combination on NSCLC and its associated characterization. METHODS AND RESULTS Antiproliferative effects were detected by cell viability assay and colony formation, and flow cytometry analysis of apoptosis and cycling showed that the combination synergistically induced apoptosis. Then, extracellular reactive oxygen species (ROS)levels and DCFH-DA-based kits examined intracellular ROS levels, and their effects on mitochondrial membrane potential were measured. Study reveals that co-induced apoptosis is associated with ROS accumulation. Subsequently, Western blotting (WB) detected the expression of epidermal growth factor receptor (EGFR), and the important signaling pathway proteins Ras/ERK and AKT/mTOR. Results showed that it could downregulation of EGFR protein expression and inhibit of activation of ERK/AKT signaling pathways. Simultaneous wound healing assay and epithelial-to-mesenchymal transition (EMT) marker detection were performed for the assessment of migration and EMT ability of NSCLC cells. Combination therapy inhibited migration and EMT of NSCLC cells. CONCLUSION The results of this study show that the combination can synergistically induce apoptosis of NSCLC by regulating ROS production. EGFR downregulation and AKT/ERK signaling pathway inhibition are linked to the synergistic effect.
Collapse
Affiliation(s)
- Huiyun Ming
- College of Basic Medical, Anhui Medical University, Hefei, 230032, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Tingting Lu
- Key Laboratory for the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, P. R. China
| | - Han Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wencheng Song
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences, Soochow University, Suzhou, 215123, China.
- Wanjiang Emerging Industry Technology Development Center, Tongling, 244000, China.
| | - Haiming Dai
- College of Basic Medical, Anhui Medical University, Hefei, 230032, China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
| |
Collapse
|
29
|
Yu Z, Zou J, Xu F. The molecular subtypes of small cell lung cancer defined by key transcription factors and their clinical significance. Lung Cancer 2024; 198:108033. [PMID: 39571251 DOI: 10.1016/j.lungcan.2024.108033] [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: 08/15/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Lung cancer, a prevalent and deadly malignancy, is classified into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). SCLC is further subdivided into four molecular subtypes-SCLC-A, SCLC-N, SCLC-P, and SCLC-I-based on key transcription factor expression. METHODS Immunohistochemistry (IHC) was used to assess ASCL1, NEUROD1, and POU2F3 expression in tumor tissues. The H-Score quantified these results. Clinical characteristics, overall survival (OS), progression-free survival (PFS), and treatment responses were analyzed by subtype, and sensitivity to different treatments was assessed. Risk factors were identified through univariate and multivariate analyses. RESULTS IHC and H-Score analysis showed that POU2F3 expression was mutually exclusive with ASCL1 or NEUROD1. Subtype distribution was as follows: SCLC-A (40 %), SCLC-N (33 %), SCLC-P (7 %), and SCLC-I (20 %). There were no significant differences in baseline characteristics, OS (p = 0.829), or PFS (p = 0.924) among subtypes. However, the SCLC-I subtype showed a trend toward improved outcomes with platinum-based doublet chemotherapy plus immune checkpoint inhibitors. Multivariate COX regression identified M stage (HR: 1.72, 95 % CI: 1.13-2.63, p = 0.012) and bone metastasis at diagnosis (HR: 1.58, 95 % CI: 1.02-2.43, p = 0.040) as independent risk factors. CONCLUSION This study confirmed the SCLC subtyping based on key transcription factors. While no significant differences in OS and PFS among subtypes were found, the SCLC-I subtype showed potential benefit from platinum-based chemotherapy combined with immune checkpoint inhibitors. M stage and bone metastasis at diagnosis were identified as independent risk factors for SCLC.
Collapse
Affiliation(s)
- Zhuchen Yu
- Clinical Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Juntao Zou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
| | - Fei Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
| |
Collapse
|
30
|
Wang H, Tang S, Wu Q, He Y, Zhu W, Xie X, Qin Z, Wang X, Zhou S, Yao S, Xu X, Guo C, Tong X, Han S, Chou YH, Wang Y, Wong KK, Yang CG, Chen L, Hu L, Ji H. Integrative study of lung cancer adeno-to-squamous transition in EGFR TKI resistance identifies RAPGEF3 as a therapeutic target. Natl Sci Rev 2024; 11:nwae392. [PMID: 39687207 PMCID: PMC11647589 DOI: 10.1093/nsr/nwae392] [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: 01/02/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 12/18/2024] Open
Abstract
Although adeno-to-squamous transition (AST) has been observed in association with resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) in clinic, its causality, molecular mechanism and overcoming strategies remain largely unclear. We here demonstrate that squamous transition occurs concomitantly with TKI resistance in PC9-derived xenograft tumors. Perturbation of squamous transition via DNp63 overexpression or knockdown leads to significant changes in TKI responses, indicative of a direct causal link between squamous transition and TKI resistance. Integrative RNA-seq, ATAC-seq analyses and functional studies reveal that FOXA1 plays an important role in maintaining adenomatous lineage and contributes to TKI sensitivity. FOXM1 overexpression together with FOXA1 knockout fully recapitulates squamous transition and TKI resistance in both PC9 xenografts and patient-derived xenograft (PDX) models. Importantly, pharmacological inhibition of RAPGEF3 combined with EGFR TKI efficiently overcomes TKI resistance, especially in RAPGEF3high PDXs. Our findings provide novel mechanistic insights into squamous transition and therapeutic strategy to overcome EGFR TKI resistance in lung cancer.
Collapse
Affiliation(s)
- Hua Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shijie Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qibiao Wu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200092, China
| | - Weikang Zhu
- Center for Excellence in Mathematical Sciences, National Center for Mathematics and Interdisciplinary Sciences, Key Laboratory of Management, Decision and Information System, Hua Loo-Keng Center for Mathematical Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Xinyun Xie
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhen Qin
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xue Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiyu Zhou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shun Yao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoling Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Chenchen Guo
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyuan Tong
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuo Han
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yueh-Hung Chou
- Department of Anatomical Pathology, Far Eastern Memorial Hospital, New Taipei City, Taiwan, China
| | - Yong Wang
- Center for Excellence in Mathematical Sciences, National Center for Mathematics and Interdisciplinary Sciences, Key Laboratory of Management, Decision and Information System, Hua Loo-Keng Center for Mathematical Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, NY 10016, USA
| | - Cai-Guang Yang
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Luonan Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Liang Hu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongbin Ji
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
31
|
Garcia NMG, Becerra JN, McKinney BJ, DiMarco AV, Wu F, Fitzgibbon M, Alvarez JV. APOBEC3 activity promotes the survival and evolution of drug-tolerant persister cells during acquired resistance to EGFR inhibitors in lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.02.547443. [PMID: 37461590 PMCID: PMC10350004 DOI: 10.1101/2023.07.02.547443] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
APOBEC mutagenesis is one of the most common endogenous sources of mutations in human cancer and is a major source of genetic intratumor heterogeneity. High levels of APOBEC mutagenesis are associated with poor prognosis and aggressive disease across diverse cancers, but the mechanistic and functional impacts of APOBEC mutagenesis on tumor evolution and therapy resistance remain relatively unexplored. To address this, we investigated the contribution of APOBEC mutagenesis to acquired therapy resistance in a model of EGFR-mutant non-small cell lung cancer. We find that inhibition of EGFR in lung cancer cells leads to a rapid and pronounced induction of APOBEC3 expression and activity. Functionally, APOBEC expression promotes the survival of drug-tolerant persister cells (DTPs) following EGFR inhibition. Constitutive expression of APOBEC3B alters the evolutionary trajectory of acquired resistance to the EGFR inhibitor gefitinib, making it more likely that resistance arises through de novo acquisition of the T790M gatekeeper mutation and squamous transdifferentiation during the DTP state. APOBEC3B expression is associated with increased expression of the squamous cell transcription factor ΔNp63 and squamous cell transdifferentiation in gefitinib-resistant cells. Knockout of p63 in gefitinib-resistant cells reduces the expression of the ΔNp63 target genes IL1α/β and sensitizes these cells to the third-generation EGFR inhibitor osimertinib. These results suggest that APOBEC activity promotes acquired resistance by facilitating evolution and transdifferentiation in DTPs, and suggest that approaches to target ΔNp63 in gefitinib-resistant lung cancers may have therapeutic benefit.
Collapse
Affiliation(s)
- Nina Marie G Garcia
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine
| | - Jessica N Becerra
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center
| | - Brock J McKinney
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center
| | - Ashley V DiMarco
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine
| | - Feinan Wu
- Genomics and Bioinformatics, Fred Hutchinson Cancer Center
| | | | - James V Alvarez
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center
| |
Collapse
|
32
|
Han X, Wang L, Wang EH. Sarcomatous transformation in metastatic EGFR-mutant lung adenocarcinoma after TKI treatment: A case report. Asian J Surg 2024; 47:4966-4967. [PMID: 38834476 DOI: 10.1016/j.asjsur.2024.05.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024] Open
Affiliation(s)
- Xu Han
- Department of Pathology, The First Hospital and the College of Basic Medical Sciences, China Medical University, Shenyang, 110001, China
| | - Liang Wang
- Department of Pathology, The First Hospital and the College of Basic Medical Sciences, China Medical University, Shenyang, 110001, China.
| | - En-Hua Wang
- Department of Pathology, The First Hospital and the College of Basic Medical Sciences, China Medical University, Shenyang, 110001, China
| |
Collapse
|
33
|
Tan N, Li Y, Ying J, Chen W. Histological transformation in lung adenocarcinoma: Insights of mechanisms and therapeutic windows. J Transl Int Med 2024; 12:452-465. [PMID: 39513032 PMCID: PMC11538883 DOI: 10.1515/jtim-2024-0019] [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] [Indexed: 11/15/2024] Open
Abstract
Histological transformation from lung adenocarcinoma (ADC) to small cell lung carcinoma (SCLC), large cell neuroendocrine carcinoma (LCNEC), squamous cell carcinoma (SCC), and sarcomatoid carcinoma (PSC) after targeted therapies is recognized as a mechanism of resistance in ADC treatments. Patients with transformed lung cancer typically experience a poor prognosis and short survival time. However, effective treatment options for these patients are currently lacking. Therefore, understanding the mechanisms underlying histological transformation is crucial for the development of effective therapies. Hypotheses including intratumoral heterogeneity, cancer stem cells, and alteration of suppressor genes have been proposed to explain the mechanism of histological transformation. In this review, we provide a comprehensive overview of the known molecular features and signaling pathways of transformed tumors, and summarized potential therapies based on previous findings.
Collapse
Affiliation(s)
- Nuopei Tan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Lin Y, Chen M, Huang S, Chen Y, Ho JH, Lin F, Tan X, Chiang H, Huang C, Tu C, Cho D, Chiu S. Targeting Dual Immune Checkpoints PD-L1 and HLA-G by Trispecific T Cell Engager for Treating Heterogeneous Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309697. [PMID: 39234811 PMCID: PMC11538689 DOI: 10.1002/advs.202309697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/04/2024] [Indexed: 09/06/2024]
Abstract
Immunotherapy targeting immune checkpoints (ICPs), such as programmed death-ligand-1 (PD-L1), is used as a treatment option for advanced or metastatic non-small cell lung cancer (NSCLC). However, overall response rate to anti-PD-L1 treatment is limited due to antigen heterogeneity and the immune-suppressive tumor microenvironment. Human leukocyte antigen-G (HLA-G), an ICP as well as a neoexpressed tumor-associated antigen, is previously demonstrated to be a beneficial target in combination with anti-PD-L1. In this study, a nanobody-based trispecific T cell engager (Nb-TriTE) is developed, capable of simultaneously binding to T cells, macrophages, and cancer cells while redirecting T cells toward tumor cells expressing PD-L1- and/or HLA-G. Nb-TriTE shows broad spectrum anti-tumor effects in vitro by augmenting cytotoxicity mediated by human peripheral blood mononuclear cells (PBMCs). In a humanized immunodeficient murine NSCLC model, Nb-TriTE exhibits superior anti-cancer potency compared to monoclonal antibodies and bispecific T cell engagers. Nb-TriTE, at the dose with pharmacoactivity, does not induce additional enhancement of circulating cytokines secretion from PMBCs. Nb-TriTE effectively prolongs the survival of mice without obvious adverse events. In conclusion, this study introduces an innovative therapeutic approach to address the challenges of immunotherapy and the tumor microenvironment in NSCLC through utilizing the dual ICP-targeting Nb-TriTE.
Collapse
Affiliation(s)
- Yu‐Chuan Lin
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
| | - Mei‐Chih Chen
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Shi‐Wei Huang
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Institute of Biomedical SciencesNational Chung Hsing UniversityTaichung City402Taiwan
| | - Yeh Chen
- Department of Food Science and BiotechnologyNational Chung Hsing UniversityTaichung City402Taiwan
| | - Jennifer Hui‐Chun Ho
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Center for Translational Genomics and Regenerative Medicine ResearchChina Medical University HospitalTaichung City404Taiwan
- Department of OphthalmologyChina Medical University HospitalChina Medical UniversityTaichung City404Taiwan
- Department of Medical ResearchEye CenterChina Medical University HospitalTaichung City404Taiwan
| | - Fang‐Yu Lin
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Xiao‐Tong Tan
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Hung‐Che Chiang
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- College of MedicineChina Medical UniversityTaichung City404Taiwan
| | - Chiu‐Ching Huang
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Division of Nephrology and the Kidney InstituteDepartment of Internal MedicineChina Medical University HospitalTaichung City404Taiwan
| | - Chih‑Yen Tu
- Division of Pulmonary and Critical CareDepartment of Internal MedicineChina Medical University HospitalTaichung City404Taiwan
- School of MedicineCollege of MedicineChina Medical UniversityTaichung City404Taiwan
| | - Der‐Yang Cho
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Drug Development CenterChina Medical UniversityTaichung City404Taiwan
- Department of NeurosurgeryChina Medical University HospitalTaichung City404Taiwan
| | - Shao‐Chih Chiu
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Drug Development CenterChina Medical UniversityTaichung City404Taiwan
| |
Collapse
|
35
|
Ladaika CA, Chakraborty A, Masood A, Hostetter G, Yi JM, O'Hagan HM. LSD1 inhibition attenuates targeted therapy-induced lineage plasticity in BRAF V600E colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620306. [PMID: 39554172 PMCID: PMC11565724 DOI: 10.1101/2024.10.25.620306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
BRAF activating mutations occur in approximately 10% of metastatic colorectal cancer (CRCs) and are associated with worse prognosis due to an inferior response to standard chemotherapy. Standard of care for patients with refractory metastatic BRAF V600E CRC is treatment with BRAF and EGFR inhibitors. However, responses are not durable. Lineage plasticity to neuroendocrine cancer is an emerging mechanism of targeted therapy resistance in several cancer types. Enteroendocrine cells (EECs), the neuroendocrine cell of the intestine, are uniquely present in BRAF V600E CRC as compared to BRAF wildtype CRC. Here, we demonstrated that combined BRAF and EGFR inhibition enriches for EECs in several models of BRAF V600E CRC. Additionally, EECs and other secretory cell types were enriched in a subset of BRAF V600E CRC patient samples following targeted therapy. Importantly, inhibition of the lysine demethylase LSD1 with a clinically relevant inhibitor attenuated targeted therapy-induced EEC enrichment through blocking the interaction of LSD1, CoREST2 and STAT3. Statement of Significance Our findings that BRAF plus EGFR inhibition induces lineage plasticity in BRAF V600E CRC represents a new paradigm for how resistance to BRAF plus EGFR inhibition occurs and our finding that LSD1 inhibition blocks lineage plasticity has the potential to improve responses to BRAF plus EGFR inhibitor therapy in patients.
Collapse
|
36
|
Shu L, Luo P, Chen Q, Liu J, Huang Y, Wu C, Pan X, Huang Z. Fibroin nanodisruptor with Ferroptosis-Autophagy synergism is potent for lung cancer treatment. Int J Pharm 2024; 664:124582. [PMID: 39142466 DOI: 10.1016/j.ijpharm.2024.124582] [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: 05/20/2024] [Revised: 07/27/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Chemotherapy agents for lung cancer often cause apoptotic resistance in cells, leading to suboptimal therapeutic outcomes. FIN56 can be a potential treatment for lung cancer as it induces non-apoptotic cell death, namely ferroptosis. However, a bottleneck exists in FIN56-induced ferroptosis treatment; specifically, FIN56 fails to induce sufficient oxidative stress and may even trigger the defense system against ferroptosis, resulting in poor therapeutic efficacy. To overcome this, this study proposed a strategy of co-delivering FIN56 and piperlongumine to enhance the ferroptosis treatment effect by increasing oxidative stress and connecting with the autophagy pathway. FIN56 and piperlongumine were encapsulated into silk fibroin-based nano-disruptors, named FP@SFN. Characterization results showed that the particle size of FP@SFN was in the nanometer range and the distribution was uniform. Both in vivo and in vitro studies demonstrated that FP@SFN could effectively eliminate A549 cells and inhibit subcutaneous lung cancer tumors. Notably, ferroptosis and autophagy were identified as the main cell death pathways through which the nano-disruptors increased oxidative stress and facilitated cell membrane rupture. In conclusion, nano-disruptors can effectively enhance the therapeutic effect of ferroptosis treatment for lung cancer through the ferroptosis-autophagy synergy mechanism, providing a reference for the development of related therapeutics.
Collapse
Affiliation(s)
- Lei Shu
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China; Panyu Central Hospital Affiliated to Guangzhou Medical University, Guangzhou 511400, PR China
| | - Peili Luo
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China
| | - Qingxin Chen
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China
| | - Jingyang Liu
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China.
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, PR China.
| |
Collapse
|
37
|
Li J, Zhan X, Shang W, Song K. Sarcomatoid carcinoma transformation in oral undifferentiated carcinoma following sequential immune combined targeted therapy: a case report. Front Immunol 2024; 15:1484915. [PMID: 39512346 PMCID: PMC11540681 DOI: 10.3389/fimmu.2024.1484915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024] Open
Abstract
The diagnosis and treatment of head and neck undifferentiated carcinoma (HNUC) present significant challenges. Herein, we present the case of a patient with advanced HNUC who underwent conversion surgery following treatment with a combination of pembrolizumab and nimotuzumab. During therapy, histological transformation from undifferentiated to sarcomatoid carcinoma was detected at the primary site. This case not only highlights the potential of immune combination-targeted therapy to reduce tumour burden and increase the surgical options for patients, but also reveals the complex alterations in tumour biology that may occur during treatment. It emphasizes the necessity for routine pathological assessments throughout the therapeutic regimen to guide personalised therapeutic strategies and optimise patient prognoses.
Collapse
Affiliation(s)
- Jieying Li
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Stomatology, Qingdao University, Qingdao, Shandong, China
| | - Xiaohong Zhan
- School of Stomatology, Qingdao University, Qingdao, Shandong, China
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei Shang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Stomatology, Qingdao University, Qingdao, Shandong, China
| | - Kai Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Stomatology, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
38
|
Gaurav I, Thakur A, Zhang K, Thakur S, Hu X, Xu Z, Kumar G, Jaganathan R, Iyaswamy A, Li M, Zhang G, Yang Z. Peptide-Conjugated Vascular Endothelial Extracellular Vesicles Encapsulating Vinorelbine for Lung Cancer Targeted Therapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1669. [PMID: 39453005 PMCID: PMC11510406 DOI: 10.3390/nano14201669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Lung cancer is one of the major cancer types and poses challenges in its treatment, including lack of specificity and harm to healthy cells. Nanoparticle-based drug delivery systems (NDDSs) show promise in overcoming these challenges. While conventional NDDSs have drawbacks, such as immune response and capture by the reticuloendothelial system (RES), extracellular vesicles (EVs) present a potential solution. EVs, which are naturally released from cells, can evade the RES without surface modification and with minimal toxicity to healthy cells. This makes them a promising candidate for developing a lung-cancer-targeting drug delivery system. EVs isolated from vascular endothelial cells, such as human umbilical endothelial-cell-derived EVs (HUVEC-EVs), have shown anti-angiogenic activity in a lung cancer mouse model; therefore, in this study, HUVEC-EVs were chosen as a carrier for drug delivery. To achieve lung-cancer-specific targeting, HUVEC-EVs were engineered to be decorated with GE11 peptides (GE11-HUVEC-EVs) via a postinsertional technique to target the epidermal growth factor receptor (EGFR) that is overexpressed on the surface of lung cancer cells. The GE11-HUVEC-EVs were loaded with vinorelbine (GE11-HUVEC-EVs-Vin), and then characterized and evaluated in in vitro and in vivo lung cancer models. Further, we examined the binding affinity of ABCB1, encoding P-glycoprotein, which plays a crucial role in chemoresistance via the efflux of the drug. Our results indicate that GE11-HUVEC-EVs-Vin effectively showed tumoricidal effects against cell and mouse models of lung cancer.
Collapse
Affiliation(s)
- Isha Gaurav
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
| | - Abhimanyu Thakur
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University (DPSRU), New Delhi 110017, India
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kui Zhang
- Ben May Department for Cancer Research, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Sudha Thakur
- National Institute for Locomotor Disabilities (Divyangjan), Kolkata 700090, India
| | - Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410017, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410017, China
| | - Gaurav Kumar
- Clinical Research Division, Department of Biosciences, School of Basic and Applied Sciences, Galgotias University, Greater Noida 203201, India
| | - Ravindran Jaganathan
- Preclinical Department, Universiti Kuala Lumpur, Royal College of Medicine Perak (UniKL-RCMP), Ipoh 30450, Malaysia
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen 518000, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
| |
Collapse
|
39
|
Oh S, Koh J, Kim TM, Kim S, Youk J, Kim M, Keam B, Jeon YK, Ku JL, Kim DW, Chung DH, Heo DS. Transcriptomic Heterogeneity of EGFR-Mutant Non-Small Cell Lung Cancer Evolution Toward Small-Cell Lung Cancer. Clin Cancer Res 2024; 30:4729-4742. [PMID: 39150541 DOI: 10.1158/1078-0432.ccr-24-0160] [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: 01/17/2024] [Revised: 06/25/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Histologic transformation from EGFR-mutant non-small cell lung cancer (NSCLC) to small-cell lung cancer (SCLC) is a key mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI). However, transcriptomic changes between NSCLC and transformed SCLC (t-SCLC) remain unexplored. EXPERIMENTAL DESIGN We conducted whole-transcriptome analysis of 59 regions of interest through the spatial profiling of formalin-fixed, paraffin-embedded tissues obtained from 10 patients (lung adenocarcinoma, 22; combined SCLC/NSCLC, 7; and t-SCLC, 30 regions of interests). Transcriptomic profiles and differentially expressed genes were compared between pre- and post-transformed tumors. RESULTS Following EGFR-TKI treatment, 93.7% (15/16) of t-SCLC components evolved into neuroendocrine-high subtypes (SCLC-A or SCLC-N). The transition to t-SCLC occurred regardless of EGFR-TKI treatment and EGFR mutational status, with a notable decrease in EGFR expression (P < 0.001) at both mRNA and protein levels. Pathway analysis revealed that gene overexpression was related to epigenetic alterations in t-SCLC. Interestingly, histone deacetylase inhibitors restored EGFR expression in SNU-2962A cells and their organoid model. The synergistic effects of third-generation EGFR-TKI osimertinib and the histone deacetylase inhibitor fimepinostat were validated in both in vitro and in vivo models. CONCLUSIONS Our study demonstrated that most t-SCLC cases showed neuronal subtypes with low EGFR expression. Differentially expressed gene analysis and t-SCLC preclinical models identified an epigenetic modifier as a promising treatment strategy for t-SCLC.
Collapse
Affiliation(s)
- Songji Oh
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Soyeon Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeonghwan Youk
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Miso Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Kyung Jeon
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Ja-Lok Ku
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong-Wan Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Doo Hyun Chung
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Dae Seog Heo
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
40
|
Dong C, Yang L, Zhao G. Circ-PGAM1 Enhances Matrine Resistance of Non-Small Cell Lung Cancer via the miR-326/CXCR5 Axis. Cancer Biother Radiopharm 2024; 39:593-599. [PMID: 36576783 DOI: 10.1089/cbr.2022.0039] [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: 12/29/2022] Open
Abstract
Background: Circular RNAs (circ-RNAs) have been demonstrated to influence initiation, drug resistance, and metastasis of tumors. However, the effects of circular-phosphoglycerate mutase 1 (circ-PGAM1) on matrine resistance in nonsmall cell lung cancer (NSCLC) remain unknown. Materials and Methods: The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to determine gene expression. The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and cell colony formation assays were used to evaluate NSCLC apoptosis and cell proliferation after indicated treatments, respectively. Results: circ-PGAM1 was upregulated in human NSCLC cell lines (H1299 and A549) compared with the human normal lung epithelial (BEAS-2B) cells. circ-PGAM1 overexpression reversed the matrine treatment-induced inhibition on proliferation of NSCLC cells (A549 and H1299) and rescued the matrine treatment-stimulated apoptosis of these cells. miR-326 was demonstrated to interact with circ-PGAM1. circ-PGAM1 knockdown enhanced the antitumor effect of matrine on NSCLC cell proliferation and apoptosis, which was reversed by miR-326 inhibition. The authors also identified CXCR5 as a key downstream target of miR-326 in A549 cells. Conclusions: circ-PGAM1 enhances matrine resistance of NSCLC cells through the miR-326/CXCR5 axis. The authors' findings provide new insights into NSCLC-targeted therapy.
Collapse
Affiliation(s)
- Caijun Dong
- Department of Thoracic Surgery, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Liangwei Yang
- Department of Cardiothoracic Surgery, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Guofang Zhao
- Department of Thoracic Surgery, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
41
|
Xiao Z, Nian Z, Zhang M, Liu Z, Zhang P, Zhang Z. Single-cell and bulk RNA-sequencing reveal SPP1 and CXCL12 as cell-to-cell communication markers to predict prognosis in lung adenocarcinoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:4610-4622. [PMID: 38622884 DOI: 10.1002/tox.24297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Lung adenocarcinoma (LUAD) generally presents as an immunosuppressive microenvironment. The characteristics of cell-to-cell communication in the LUAD microenvironment has been unclear. In this study, the LUAD bulk RNA-seq data and single-cell RNA-seq data were retrieved from public dataset. Differential expression genes (DEGs) between LUAD tumor and adjacent non-tumor tissues were calculated by limma algorithm, and then detected by PPI, KEGG, and GO analysis. Cell-cell interactions were explored using the single-cell RNA-seq data. Finally, the first 15 CytoHubba genes were used to establish related pathways and these pathways were used to characterize the immune-related ligands and their receptors in LUAD. Our analyses showed that monocytes or macrophages interact with tissue stem cells and NK cells via SPP1 signaling pathway and tissue stem cells interact with T and B cells via CXCL signaling pathway in different states. Hub genes of SPP1 participated in SPP1 signaling pathway, which was negatively correlated with CD4+ T cell and CD8+ T cell. The expression of SPP1 in LUAD tumor tissues was negatively correlated with the prognosis. While CXCL12 participated in CXCL signaling pathway, which was positively correlated with CD4+ T cell and CD8+ T cell. The role of CXCL12 in LUAD tumor tissues exhibits an opposite effect to that of SPP1. This study reveals that tumor-associated monocytes or macrophages may affect tumor progression. Moreover, the SPP1 and CXCL12 may be the critic genes of cell-to-cell communication in LUAD, and targeting these pathways may provide a new molecular mechanism for the treatment of LUAD.
Collapse
Affiliation(s)
- Zengtuan Xiao
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
- Department of Immunology, Biochemistry and Molecular Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China
| | - Zhe Nian
- Department of Immunology, Biochemistry and Molecular Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Zuo Liu
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| |
Collapse
|
42
|
Sun X, Kang T, Liu B, Zhang Y, Huang G. A Meta-Analysis of Efficacy and Safety of Neoadjuvant Immunotherapy Plus Chemotherapy for Resectable Non-Small Cell Lung Cancer. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70019. [PMID: 39359047 PMCID: PMC11447246 DOI: 10.1111/crj.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
INTRODUCTION Neoadjuvant immunotherapy plus chemotherapy has ushered in a new era for surgical treatment for patients with NSCLC. This study aimed to examine the efficacy and safety of neoadjuvant immunotherapy plus chemotherapy in NSCLC. METHODS Eligible studies were identified from PubMed, Embase, Web of Science, Cochrane Library, ClinicalTrials.gov, and conference meeting abstracts. The endpoints included major pathological response (MPR), complete pathological response (pCR), surgical resection rate, R0 resection, treatment-related adverse events (TRAEs), severe adverse events (SAEs), surgical complications, treatment discontinuation, surgical delay, and treatment-related death. Stata 18 software was used for statistical analysis, and p < 0.05 was considered statistically significant. Twenty-two studies including a total of 1108 patients were eligible for this study. RESULTS Among the patients who received neoadjuvant immunotherapy plus chemotherapy, the pooled MPR rate was 51% (95% CI [0.44-0.58]), and pCR rate was 34% (95% CI [0.28-0.40]). The pooled surgical resection rate was 85% (95% CI [0.81-0.89]), and the pooled R0 rate was 94% (95% CI [0.91-0.96]). The pooled rate of pathological tumor downstaging was 84% (95% CI [0.79-0.88]), and the pooled rate of pathological nodal downstaging was 38% (95% CI [0.23-0.57]). During the treatment of neoadjuvant immunotherapy plus chemotherapy with or without surgery, the pooled rate of TRAEs (any grade) was 84% (95% CI [0.73-0.91]), and the pooled rate of SAEs was 29% (95% CI [0.21-0.38]). Surgical complications pooled rate was 25% (95% CI [0.14-0.41]). The pooled rate of treatment discontinuation (11%, 95% CI [0.09-0.13]), surgical delay (3%, 95% CI [0.02-0.05]), and treatment-related death (2%, 95% CI [0.02-0.03]) were conducted. CONCLUSION Neoadjuvant immunotherapy plus chemotherapy provides a high pathological response, surgical resection rate, R0 resection rate, and pathological downstage rate and has a low risk of increasing the incidence of SAEs, surgical complications, treatment discontinuation, surgical delay, and treatment-related death. The validation of prospective and large sample studies is needed to confirm this conclusion.
Collapse
Affiliation(s)
- Xinru Sun
- Department of Interventional OncologyZibo Central HospitalZiboShandong ProvinceChina
| | - Tianhua Kang
- Department of Interventional OncologyZibo Central HospitalZiboShandong ProvinceChina
| | - Baodong Liu
- Department of Interventional OncologyZibo Central HospitalZiboShandong ProvinceChina
| | - Yin Zhang
- Department of Interventional OncologyZibo Central HospitalZiboShandong ProvinceChina
| | - Guangming Huang
- Department of Interventional OncologyZibo Central HospitalZiboShandong ProvinceChina
| |
Collapse
|
43
|
Lyu HX, Ma WH, Zhang YQ, Jin H, Wang YD, Zhao M. Case report: Emerging therapies for transformed small cell lung cancer: efficacy of serplulimab and a comprehensive case report. Front Med (Lausanne) 2024; 11:1406515. [PMID: 39386753 PMCID: PMC11461193 DOI: 10.3389/fmed.2024.1406515] [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: 03/25/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
This research reports a case of histological transformation from non-small cell lung cancer (NSCLC) to transformed small cell lung cancer (T-SCLC) in a patient undergoing EGFR-tyrosine kinase inhibitors (TKIs). The aggressive characteristics of the tumor diverged significantly from those commonly associated with lung adenocarcinomas, leading to further histological analysis. The subsequent histological examination confirmed the transformation to SCLC, consistent with established mechanisms of acquired resistance in NSCLC. Given the limited therapeutic options, the patient was administered a serplulimab-based immunochemotherapy regimen, achieving a progression-free survival (PFS) of 6 months post-transformation. The study underscores the potential of PD-1 inhibitors, particularly serplulimab, in the treatment landscape for T-SCLC and highlights the need for future comprehensive research.
Collapse
Affiliation(s)
- Heng-Xu Lyu
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wen-Hua Ma
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong-Qian Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hui Jin
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yu-Dong Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Zhao
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
44
|
An J, Zhang M, Fu Y, Zhang Q, Si Y, Zhang Y, Fang Y, Zhang D. Emerging electrochemical biosensors for lung cancer-associated protein biomarker and miRNA detection. Int J Biol Macromol 2024; 280:135972. [PMID: 39322139 DOI: 10.1016/j.ijbiomac.2024.135972] [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: 06/06/2024] [Revised: 09/09/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Lung cancer remains a major driver of global morbidity and mortality, and diagnosing lung tumors early in their development is vital to maximizing treatment efficacy and patient survival. Several biomarkers, including CYFRA 21-1, NSE, ProGRP, CEA, and miRNA, have been identified as reliable indicators for early lung cancer detection and monitoring treatment progress. However, the minute changes in the levels of these biomarkers during the early stages of disease necessitate advanced detection platforms. In this space, electrochemical biosensors have currently emerged as robust tools for early lung cancer screening and diagnosis owing to their low costs, rapid responses, and superior sensitivity and selectivity. This review provides an up-to-date overview of the application of electrochemiluminescence, photoelectrochemical, and other electrochemical analytical strategies for detecting lung cancer-associated protein biomarkers, and miRNA. This review compares these techniques to provide a concise overview of the principles underlying these electrochemical analytical methods, the preparation of their components, and the performance of the resulting biosensors. Lastly, a discussion of the challenges and opportunities associated with electrochemical biosensors detection of lung cancer-associated biomarkers are provided.
Collapse
Affiliation(s)
- Jiaying An
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Miao Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yu Fu
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qingxiang Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yuxin Si
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Youlin Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, PR China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China.
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China.
| |
Collapse
|
45
|
Lee CY, Wu YC, Liao TC, Hsiao SH, Hsu JBK, Chang TH. A Study of Disease Prognosis in Lung Adenocarcinoma Using Single-Cell Decomposition and Immune Signature Analysis. Cancers (Basel) 2024; 16:3207. [PMID: 39335178 PMCID: PMC11431002 DOI: 10.3390/cancers16183207] [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: 08/05/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The development of tumors is a highly complex process that entails numerous interactions and intricate relationships between the host immune system and cancer cells. It has been demonstrated in studies that the treatment response of patients can be correlated with the tumor microenvironment (TME). Consequently, the examination of diverse immune profiles within the TME can facilitate the elucidation of tumor development and the development of advantageous models for diagnoses and prognoses. Methods: In this study, we utilized a single-cell decomposition method to analyze the relationships between cell proportions and immune signatures in lung adenocarcinoma (LUAD) patients. Results: Our findings indicate that specific immune cell populations and immune signatures are significantly associated with patient prognosis. By identifying poor prognosis signatures (PPS), we reveal the critical role of immune profiles and cellular composition in disease outcomes, emphasizing their diagnostic potential for predicting patient prognosis. Conclusions: This study highlights the importance of immune signatures and cellular composition, which may serve as valuable biomarkers for disease prognosis in LUAD patients.
Collapse
Affiliation(s)
- Cheng-Yang Lee
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; (C.-Y.L.); (T.-C.L.)
| | - Yu-Chung Wu
- Division of Thoracic Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110301, Taiwan;
- Department of Surgery, Division of Thoracic Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Tze-Chi Liao
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; (C.-Y.L.); (T.-C.L.)
| | - Shih-Hsin Hsiao
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Justin Bo-Kai Hsu
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; (C.-Y.L.); (T.-C.L.)
| |
Collapse
|
46
|
Forbes AN, Xu D, Cohen S, Pancholi P, Khurana E. Discovery of therapeutic targets in cancer using chromatin accessibility and transcriptomic data. Cell Syst 2024; 15:824-837.e6. [PMID: 39236711 PMCID: PMC11415227 DOI: 10.1016/j.cels.2024.08.004] [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: 01/07/2022] [Revised: 09/22/2023] [Accepted: 08/08/2024] [Indexed: 09/07/2024]
Abstract
Most cancer types lack targeted therapeutic options, and when first-line targeted therapies are available, treatment resistance is a huge challenge. Recent technological advances enable the use of assay for transposase-accessible chromatin with sequencing (ATAC-seq) and RNA sequencing (RNA-seq) on patient tissue in a high-throughput manner. Here, we present a computational approach that leverages these datasets to identify drug targets based on tumor lineage. We constructed gene regulatory networks for 371 patients of 22 cancer types using machine learning approaches trained with three-dimensional genomic data for enhancer-to-promoter contacts. Next, we identified the key transcription factors (TFs) in these networks, which are used to find therapeutic vulnerabilities, by direct targeting of either TFs or the proteins that they interact with. We validated four candidates identified for neuroendocrine, liver, and renal cancers, which have a dismal prognosis with current therapeutic options.
Collapse
Affiliation(s)
- Andre Neil Forbes
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sandra Cohen
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Priya Pancholi
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
47
|
Niu X, Liu W, Zhang Y, Liu J, Zhang J, Li B, Qiu Y, Zhao P, Wang Z, Wang Z. Cancer plasticity in therapy resistance: Mechanisms and novel strategies. Drug Resist Updat 2024; 76:101114. [PMID: 38924995 DOI: 10.1016/j.drup.2024.101114] [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: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Therapy resistance poses a significant obstacle to effective cancer treatment. Recent insights into cell plasticity as a new paradigm for understanding resistance to treatment: as cancer progresses, cancer cells experience phenotypic and molecular alterations, corporately known as cell plasticity. These alterations are caused by microenvironment factors, stochastic genetic and epigenetic changes, and/or selective pressure engendered by treatment, resulting in tumor heterogeneity and therapy resistance. Increasing evidence suggests that cancer cells display remarkable intrinsic plasticity and reversibly adapt to dynamic microenvironment conditions. Dynamic interactions between cell states and with the surrounding microenvironment form a flexible tumor ecosystem, which is able to quickly adapt to external pressure, especially treatment. Here, this review delineates the formation of cancer cell plasticity (CCP) as well as its manipulation of cancer escape from treatment. Furthermore, the intrinsic and extrinsic mechanisms driving CCP that promote the development of therapy resistance is summarized. Novel treatment strategies, e.g., inhibiting or reversing CCP is also proposed. Moreover, the review discusses the multiple lines of ongoing clinical trials globally aimed at ameliorating therapy resistance. Such advances provide directions for the development of new treatment modalities and combination therapies against CCP in the context of therapy resistance.
Collapse
Affiliation(s)
- Xing Niu
- China Medical University, Shenyang, Liaoning 110122, China; Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, 999077, Hong Kong, China
| | - Wenjing Liu
- Medical Oncology Department of Thoracic Cancer (2), Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yinling Zhang
- Department of Oncology Radiotherapy 1, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong 266042, China
| | - Jing Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Bo Li
- Department of Orthopedics, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Peng Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Zhongmiao Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Zhe Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
48
|
Wang Q, Liu XY, Zhang XQ, Huo ZX, Chen CY, Chen S, Liu CY, Zhu J, Liu SS, Lu B. LRRC45 promotes lung cancer proliferation and progression by enhancing c-MYC, slug, MMP2, and MMP9 expression. Adv Med Sci 2024; 69:451-462. [PMID: 39326735 DOI: 10.1016/j.advms.2024.09.007] [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: 08/17/2023] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The leucine-rich repeat-containing (LRRC) superfamily members are known for their significant roles in tumorigenesis and cellular proliferation. However, the specific regulatory role of LRRC45 in lung cancer remains unexplored. This study investigated the impact and underlying mechanisms of LRRC45 on the proliferative, migratory, and invasive capacities of lung adenocarcinoma (LUAD) cells, potentially identifying new targets for therapeutic intervention. MATERIAL AND METHODS The importance of LRRC45 in lung cancer was analyzed using the online databases of UCSC Xena, TCGA, TISIDB, and UALCAN, whereas to detect target gene expression, we used the qRT-PCR, Western blot, and immunofluorescence confocal. The cell growth was monitored by colony formation assay and migration was examined by cell migration assay. Finally, a xenograft mouse tumor model using A549 cells was used to explore the in vivo effect of LRRC45 in lung cancer. RESULTS Inhibition of LRRC45 expression led to a notable decrease in proliferation, migration, and invasion of A549 and H1299 cells. LRRC45 silencing significantly reduced the tumor volume and improved the mice's survival. Additionally, inhibition of LRRC45 expression dramatically suppressed c-MYC, Slug, MMP2, and MMP9 expression. Overexpression of c-MYC and/or Slug in the LRRC45-deficient cells can partially or totally restore the LRRC45 deficiency-suppressed growth. Moreover, the overexpression of MMP2 and/or MMP9 could partially or totally restore LRRC45 deficiency-reduced cell metastasis. CONCLUSIONS LRRC45 could promote the proliferative, migrative, and invasive capacities of lung cancer cells by increasing c-MYC, Slug, MMP2, and MMP9 expression, indicating the therapeutic implications and potential significance of these pathways in lung cancer.
Collapse
Affiliation(s)
- Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China; Department of Respiratory Medicine, Suqian Affiliated Hospital of Nanjing University of Chinese Medicine, Suqian Hospital of Chinese Medicine, Suqian, Jiangsu, China.
| | - Xin-Yan Liu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiao-Qi Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zheng-Xing Huo
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Cheng-Yu Chen
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shi Chen
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Cheng-Yong Liu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jia Zhu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shan-Shan Liu
- Department of Respiratory Medicine, Suqian Affiliated Hospital of Nanjing University of Chinese Medicine, Suqian Hospital of Chinese Medicine, Suqian, Jiangsu, China.
| | - Bing Lu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Taicang Hospital of Traditional Chinese Medicine, Taicang, Jiangsu, China.
| |
Collapse
|
49
|
Zhou Q, Ye Z, Xu X, Zhong Y, Luo J, Zhang Z, Chen J, Chen Z, Cai J, Zhang X, Qian J. Drug-induced enzyme activity inhibition and CYP3A4 genetic polymorphism significantly shape the metabolic characteristics of furmonertinib. Toxicology 2024; 507:153903. [PMID: 39098371 DOI: 10.1016/j.tox.2024.153903] [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: 05/25/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/06/2024]
Abstract
This study aimed to elucidate the impact of variations in liver enzyme activity, particularly CYP3A4, on the metabolism of furmonertinib. An in vitro enzyme incubation system was established for furmonertinib using liver microsomes and recombinant CYP3A4 baculosomes, with analytes detected by LC-MS/MS. The pharmacokinetic characteristics of furmonertinib were studied in vivo using Sprague-Dawley rats. It was found that telmisartan significantly inhibited the metabolism of furmonertinib, as demonstrated by a significant increase in the AUC of furmonertinib when co-administered with telmisartan, compared to the furmonertinib-alone group. Mechanistically, it was noncompetitive in rat liver microsomes, while it was mixed competitive and noncompetitive in human liver microsomes and CYP3A4. Considering the genetic polymorphism of CYP3A4, the study further investigated its effect on the kinetics of furmonertinib. The results showed that compared to CYP3A4.1, CYP3A4.29 had significantly increased activity in catalyzing furmonertinib, whereas CYP3A4.7, 9, 10, 12, 13, 14, 18, 23, 33, and 34 showed markedly decreased activity. The inhibitory activity of telmisartan varied in CYP3A4.1 and CYP3A4.18, with IC50 values of 8.56 ± 0.90 μM and 27.48 ± 3.52 μM, respectively. The key loci affecting the inhibitory effect were identified as ARG105, ILE301, ALA370, and LEU373. Collectively, these data would provide a reference for the quantitative application of furmonertinib.
Collapse
Affiliation(s)
- Qi Zhou
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhize Ye
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, China
| | - Xiaoyu Xu
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunshan Zhong
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianchao Luo
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheyan Zhang
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Chen
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongxi Chen
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianping Cai
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China.
| | | | - Jianchang Qian
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
50
|
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
|