1
|
Li J, Gao X, Lv L, Huang Y, Zhang H, Sun X, Zhu L. Development of a coagulation‑related gene model for prognostication, immune response and treatment prediction in lung adenocarcinoma. Oncol Lett 2025; 29:290. [PMID: 40276086 PMCID: PMC12018795 DOI: 10.3892/ol.2025.15035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/13/2025] [Indexed: 04/26/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is the most prevalent form of lung cancer worldwide. Due to the lack of clinically useful molecular biomarkers, the diagnosis and prognosis of patients with LUAD remain poor. Patients with LUAD often exhibit abnormalities in the levels of coagulation factors. Therefore, the objective of the present study was to develop a model based on coagulation-related factors in LUAD. Gene expression data and clinical information from 582 patients with LUAD were obtained from The Cancer Genome Atlas (TCGA). A set of 138 coagulation-related genes (CRGs) was retrieved from The Molecular Signatures Database, and their expression levels were examined in TCGA dataset to identify differentially expressed CRGs. Predictive models were constructed using least absolute shrinkage and selection operator-Cox regression. The risk score from the model was used to establish high- and low-risk patient groups. Additionally, Kaplan-Meier analyses were performed to evaluate the differences in overall survival (OS) and progression-free survival between the two groups. The accuracy of the model was verified through receiver operating characteristic and principal component analysis. In addition, the tumor immune dysfunction and exclusion algorithm was used to assess immune escape and immunotherapy responses in relation to the CRGs. A predictive model comprising four genes, namely matrix metalloproteinase (MMP) 1, MMP10, cathepsin V and thrombin, was established to estimate the survival rate of patients with LUAD. The OS rates of patients in the high-risk group were lower compared with those in the low-risk group. Furthermore, a combination of high-risk score and low tumor mutation burden was associated with the poorest survival in patients with LUAD. Patients in different risk groups exhibited different drug sensitivities based on their risk scores. In conclusion, the four-gene based prognostic model served as an independent predictor of survival rates in patients with LUAD and may offer a novel approach for prognosis and treatment.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100000, P.R. China
- Department of Cardiac Surgery, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100000, P.R. China
| | - Xuedi Gao
- Department of Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong 250013, P.R. China
| | - Lin Lv
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Yubin Huang
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Houlu Zhang
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Xiaoming Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Liangming Zhu
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
- Department of Thoracic Surgery, Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
2
|
Zhou G, Tan J, Zhang P, Zhou Z, Zhang L, Zhang Z. Mechanistic Insights and Therapeutic Potentials of Ubiquitin-Proteasome System in Non-Small Cell Lung Cancer. Cell Prolif 2025:e70050. [PMID: 40313038 DOI: 10.1111/cpr.70050] [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: 03/08/2025] [Revised: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 05/03/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer mortality. Despite advancements in gene targeted therapies and immunotherapies, high heterogeneity contributes to limited efficacy and therapeutic resistance. Ubiquitination, a crucial post-translational modification that regulates protein stability and degradation, plays a significant role in cancer pathogenesis by influencing key oncogenic pathways and tumour progression. This review systematically explores the ubiquitin-proteasome system (UPS) and its potential as a therapeutic target for NSCLC. We highlight recent preclinical and clinical studies focusing on ubiquitination-related biomarkers, drug targets and emerging therapies like proteasome inhibitors and Proteolysis-targeting chimeras (PROTACs). By exploring the impact of the UPS on tumour biology, the progression of NSCLC and its response to therapy, we aim to underscore the potential of targeting the ubiquitination-deubiquitination system as a complementary or synergistic approach to existing therapeutic strategies in NSCLC, thereby enhancing patient outcomes and overcoming treatment resistance.
Collapse
Affiliation(s)
- Guangyao Zhou
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jiaxiong Tan
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lianmin Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
3
|
Tiwade PB, Fung V, VanKeulen-Miller R, Narasipura EA, Ma Y, Fenton OS. Non-Viral RNA Therapies for Non-Small Cell Lung Cancer and Their Corresponding Clinical Trials. Mol Pharm 2025; 22:1752-1774. [PMID: 40131145 DOI: 10.1021/acs.molpharmaceut.4c00871] [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: 03/26/2025]
Abstract
Ribonucleic acid (RNA)-based therapies represent a promising class of drugs for the treatment of non-small cell lung cancer (NSCLC) due to their ability to modulate gene expression. Therapies leveraging small interfering RNA (siRNA), messenger RNA (mRNA), microRNA (miRNA), and antisense oligonucleotides (ASOs) offer various advantages over conventional treatments, including the ability to target specific genetic mutations and the potential for personalized medicine approaches. However, the clinical translation of these therapeutics for the treatment of NSCLC faces challenges in delivery due to their immunogenicity, negative charge, and large size, which can be mitigated with delivery platforms. In this review, we provide a description of the pathophysiology of NSCLC and an overview of RNA-based therapeutics, specifically highlighting their potential application in the treatment of NSCLC. We discuss relevant classes of RNA and their therapeutic potential for NSCLC. We then discuss challenges in delivery and non-viral delivery strategies such as lipid- and polymer-based nanoparticles that have been developed to address these issues in preclinical models. Furthermore, we provide a summary table of clinical trials that leverage RNA therapies for NSCLC [which includes their National Clinical Trial (NCT) numbers] to highlight the current progress in NSCLC. We also discuss how these NSCLC therapies can be integrated with existing treatment modalities to enhance their efficacy and improve patient outcomes. Overall, we aim to highlight non-viral strategies that tackle RNA delivery challenges while showcasing RNA's potential as a next-generation therapy for NSCLC treatment.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/therapy
- Lung Neoplasms/drug therapy
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- RNA, Small Interfering/administration & dosage
- Oligonucleotides, Antisense/therapeutic use
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/administration & dosage
- Clinical Trials as Topic
- Animals
- Nanoparticles/chemistry
- MicroRNAs/genetics
- MicroRNAs/therapeutic use
- RNA, Messenger/genetics
- Genetic Therapy/methods
- Drug Delivery Systems/methods
Collapse
Affiliation(s)
- Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eshan Amruth Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
4
|
Li S, Hu Y, Tian C, Luan J, Zhang X, Wei Q, Li X, Bian Y. Prediction of EGFR-TP53 genes co-mutations in patients with lung adenocarcinoma (LUAD) by 18F-FDG PET/CT radiomics. Clin Transl Oncol 2025; 27:1506-1515. [PMID: 39251494 DOI: 10.1007/s12094-024-03685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE This retrospective study was undertaken to assess the predictive efficacy of 18F-FDG PET/CT -derived radiomic features concerning the co-mutation status of epidermal growth factor receptor (EGFR) and TP53 in LUAD. METHODS A cohort of 150 LUAD patients underwent pretreatment 18F-FDG PET/CT scans with known mutation status of EGFR and TP53 were collected. The feature extraction based on their PET/CT images utilized the Pyradiomics package based on the 3D Slicer. The optimal radiomic features were selected through correlation analysis and the Gradient Boosting Decision Tree (GBDT) algorithm, followed by the construction of the radiomic model. The clinical model incorporated meaningful clinical variables, whereas the complex model integrated both the radiomic and clinical models. The area under the receiver operating characteristic curve (AUC) facilitated the comparison of prediction performance across the three models. The DCA gauged the clinical utility of these models. RESULTS The patient cohort was randomly allocated into a training set (n = 105) and a validation set (n = 45) in a 7:3 ratio. Eleven PET and eleven CT optimal radiomic features were selected to construct the radiomic model. The model showed a good ability to discriminate the co-occurrence of EGFR and TP53, with AUC equal to 0.850 in the training set, and 0.748 in the validation set, compared with 0.750 and 0.626 for the clinical model. The complex model exhibited the highest AUC values, with 0.880 and 0.794 in both sets, but there were no significant differences compared to the radiomic model. The DCA revealed favorable clinical value. CONCLUSION
Collapse
Affiliation(s)
- Shuheng Li
- Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yujing Hu
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Congna Tian
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jiusong Luan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qiang Wei
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaodong Li
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yanzhu Bian
- Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
5
|
Su Y, Huo T, Wang Y, Li J. Construction and clinical significance of prognostic risk markers based on cancer driver genes in lung adenocarcinoma. Clin Transl Oncol 2025; 27:1539-1557. [PMID: 39292390 DOI: 10.1007/s12094-024-03703-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Cancer driver genes (CDGs) have been reported as key factors influencing the progression of lung adenocarcinoma (LUAD). However, the role of CDGs in LUAD prognosis has not been fully elucidated. METHODS LUAD transcriptome data and CDG-related data were obtained from public databases and literature. Differentially expressed CDGs (DE-CDGs) greatly associated with LUAD survival (P < 0.05) were identified to establish a prognostic model. In addition, immune analysis of high-risk (HR) and low-risk (LR) groups was conducted by utilizing the CIBERSORT and single sample gene set enrichment analysis (ssGSEA) algorithms to assess immune differences. Subsequently, mutation analysis was conducted using maftools. Finally, candidate drugs were identified using the CellMiner database. RESULTS 40 DE-CDGs significantly associated with LUAD survival and 11 DE-CDGs associated with prognosis were identified through screening. Regression analysis revealed that risk score can independently predict LUAD prognosis (P < 0.05). Immune landscape analysis revealed that compared to the HR group, the LR group had higher immune scores and high infiltration of various immune cells such as follicular helper B cells and T cells. Mutation landscape analysis demonstrated that missense mutation was the most common mutation type in both risk groups. Drug prediction analysis revealed strong correlations of fulvestrant, S-63845, sapacitabine, lomustine, BLU-667, SR16157, motesanib, AZD-9496, XK-469, dimethylfasudil, P-529, and imatinib with the model genes, suggesting their potential as candidate drugs targeting the model genes. CONCLUSION This study identified 11 effective biomarkers, DE-CDGs, which can predict LUAD prognosis and explored the biological significance of CDGs in LUAD prognosis, immunotherapy, and treatment.
Collapse
Affiliation(s)
- Yazhou Su
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui, 453100, Henan province, China.
| | - Tingting Huo
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan province, China
| | - Yanan Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan province, China
| | - Jingyan Li
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan province, China
| |
Collapse
|
6
|
Bao ZC, Zhang Y, Liu ZD, Dai HJ, Ren F, Li N, Lv SY, Zhang Y. Tetrahydrocurcumin-induced apoptosis of hepatocellular carcinoma cells involves the TP53 signaling pathway, as determined by network pharmacology. World J Gastrointest Oncol 2025; 17:101174. [PMID: 40092919 PMCID: PMC11866214 DOI: 10.4251/wjgo.v17.i3.101174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/04/2024] [Accepted: 01/16/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant disease with high incidence and mortality worldwide. This study focuses on the TP53 target protein to investigate the potential therapeutic effect of tetrahydrocurcumin (THC) on HCC and its mechanism of action. The research hypothesis is that THC can inhibit the proliferation, migration, and invasion of HCC cells, and promote their apoptosis by regulating the TP53 target protein. AIM To explore the mechanism by which THC inhibits HCC cell proliferation via the TP53 signaling pathway. METHODS Potential targets of THC and HCC were identified from multiple databases. The core targets were subjected to analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases, and visualization processing, using the online platform Metascape to identify the key molecules and signaling pathways involved in the action of THC against HCC. The molecular mechanisms of action of THC against TP53 in the inhibition of HCC cells were verified using cell counting kit-8, Transwell, apoptosis, and western blotting assays. RESULTS Molecular docking results showed that THC had a high score for the TP53 target protein. In vitro experiments indicated that THC effectively inhibited the proliferation and migration of HCC cells, and affected the expression levels of TP53, MDM2, cyclin B, Bax, Bcl-2, caspase-9, and caspase-3. CONCLUSION THC induces the apoptosis of HCC cells through the TP53 signaling pathway, thereby inhibiting their proliferation and migration.
Collapse
Affiliation(s)
- Zhuo-Cong Bao
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ye Zhang
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Zhao-Dong Liu
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Hui-Jun Dai
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Fu Ren
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ning Li
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Shang-Yu Lv
- Department of Clinical Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yan Zhang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- International Education School, International Exchange and Cooperation Office, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| |
Collapse
|
7
|
Du F, Zhang A, Qi X, Yin R, Jiang T, Li J. Novel Camptothecin Derivative 9c with Enhanced Antitumor Activity via NSA2-EGFR-P53 Signaling Pathway. Int J Mol Sci 2025; 26:1987. [PMID: 40076615 PMCID: PMC11900506 DOI: 10.3390/ijms26051987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Therapeutic challenges persist in the management of non-small cell lung cancer (NSCLC) in oncology. Camptothecins have demonstrated as crucial agents in tumor therapy; however, their efficacy is significantly hindered by adverse effects and drug resistance. Herein, we present a novel camptothecin derivative named 9c, which exhibits impressive anti-NSCLC potency surpassing the widely recognized camptothecin analog FL118 through a novel mechanism. Our findings demonstrated that 9c effectively inhibited tumor malignancy through cell cycle arrest and apoptosis induction with the transcriptional downregulation of anti-apoptotic genes including survivin, Mcl-1, Bcl-2, and XIAP. Mechanistically, 9c induced a wild-type p53 expression by destabilizing the NSA2-EGFR axis, thus delaying the cell cycle progression and ultimately triggering apoptosis. 9c significantly inhibited the growth of the NSCLC xenograft in vivo without observed side toxicity. Importantly, it complemented the therapeutic advantages of the novel drug AMG510 for addressing KRAS-mutant NSCLC. Collectively, these findings position 9c as a promising candidate with innovative approaches to combat NSCLC.
Collapse
Affiliation(s)
- Fu Du
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Aotong Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Xin Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Ruijuan Yin
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| |
Collapse
|
8
|
Dong Y, Yang G, Yang Y, Zhang S, Wang Y, Xu H. Dynamic characterization of circulating tumor DNA in HER2-altered advanced non-small cell lung cancer treated with pyrotinib and apatinib: Exploratory biomarker analysis from PATHER2 study. Lung Cancer 2025; 200:108062. [PMID: 39827483 DOI: 10.1016/j.lungcan.2024.108062] [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: 07/04/2024] [Revised: 12/01/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND HER2 mutations are critical drivers of non-small cell lung cancer (NSCLC), affecting 2 %-3 % of patients and often leads to poor prognosis and limited response to conventional therapies. This study investigates the genomic characteristics and prognostic relevance of dynamic circulating tumor DNA (ctDNA) monitoring in advanced NSCLC patients with HER2 mutations treated with pyrotinib and apatinib. METHODS The PATHER2 study included 33 advanced NSCLC patients harboring HER2 mutations or amplification, who received combination therapy of pyrotinib and apatinib. Among them, 27 patients had baseline blood samples available for analysis. Baseline blood samples (n = 27), follow-up samples after one treatment cycle (n = 13), and samples upon disease progression (n = 18) were collected. ctDNA was extracted and sequenced using a 556-gene panel. RESULTS At baseline, HER2 mutations were detected in 21 of 27 patients through ctDNA, and 19 showed consistent results between tissue and blood sample testing. Patients with TP53 and DNMT3A alterations at baseline had significantly shorter progression-free survival (PFS). Dynamic ctDNA monitoring revealed that patients without detectable HER2 mutations after one treatment cycle had longer PFS and a trend toward longer overall survival (OS) compared to those with persistent HER2 mutations. The newly emerged mutations after resistance were infrequently found in HER2, instead primarily enriched in the chromatin remodeling pathway. CONCLUSION ctDNA holds significant value in guiding the treatment of patients with HER2 mutations. Baseline TP53 and DNMT3A alterations, along with persistent HER2 mutations after initial treatment, are associated with poorer prognosis. The primary mechanism of resistance to pyrotinib and apatinib in these patients may be attributed to chromatin remodeling rather than on-target alterations.
Collapse
Affiliation(s)
- Yucheng Dong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guangjian Yang
- Department of Respiratory Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yaning Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuyang Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Haiyan Xu
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
9
|
Ye Z, Huang T, Hu K, Zhou H, Huang L, Wang L. Genomic Profiling Reveals Immune-Related Gene Differences in Lung Cancer Patients Stratified by PD1/PDL1 Expression: Implications for Immunotherapy Efficacy. J Appl Genet 2025; 66:105-114. [PMID: 38363451 DOI: 10.1007/s13353-024-00841-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Lung cancer remains a leading cause of global cancer-related mortality, and the exploration of innovative therapeutic approaches, such as PD1/PDL1 immunotherapy, is critical. This study leverages comprehensive data from the Cancer Genome Atlas (TCGA) to investigate the differential expression of PD1/PDL1 in lung cancer patients and explores its implications. Clinical data, RNA expression, somatic mutations, and copy number variations of 1017 lung cancer patients were obtained from TCGA. Patients were categorized into high (HE) and low (LE) PD1/PDL1 expression groups based on mRNA levels. Analyses included differential gene expression, functional enrichment, protein-protein interaction networks, and mutational landscape exploration. The study identified 391 differentially expressed genes, with CD4 and PTPRC among the upregulated genes in the HE group. Although overall survival did not significantly differ between HE and LE groups, enrichment analysis revealed a strong association with immunoregulatory signaling pathways, emphasizing the relevance of PD1/PDL1 in immune response modulation. Notably, TP53 mutations were significantly correlated with high PD1/PDL1 expression. This study provides a comprehensive analysis of PD1/PDL1 expression in lung cancer, uncovering potential biomarkers and highlighting the intricate interplay between PD1/PDL1 and the immune response. The identified upregulated genes, including CD4 and PTPRC, warrant further investigation for their roles in the context of lung cancer and immunotherapy. The study underscores the importance of considering molecular heterogeneity in shaping personalized treatment strategies for lung cancer patients. Limitations, such as the retrospective nature of TCGA data, should be acknowledged.
Collapse
Affiliation(s)
- Zhifeng Ye
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Ting Huang
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Keke Hu
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - HeRan Zhou
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Ling Huang
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Lu Wang
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China.
| |
Collapse
|
10
|
Wan X, Zhang C, Kang M, Rossi A, Goto T, Seetharamu N, Seki N, Lu H, Zhang Y. Analysis and exploration of regulatory mechanisms and potential prognostic biomarkers in squamous cell carcinoma of the lung by expression profiling. Transl Cancer Res 2025; 14:569-583. [PMID: 39974402 PMCID: PMC11833388 DOI: 10.21037/tcr-2024-2443] [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: 12/03/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Background Lung cancer is the most common malignant tumor in China. In 2016, more than 800,000 new cases of lung cancer were diagnosed in China. Squamous cell carcinoma of the lung, a type of non-small cell lung cancer (NSCLC), accounts for 25-30% of all lung cancer cases, and has an overall 5-year survival rate of about 32.53%, lower than adenocarcinoma for which there have been far more therapeutic advances in the last few decades. The purpose of this study was to explore the mechanisms of the disease and to identify potential prognostic biomarkers. Methods This study analyzed lung squamous cell carcinoma of the lung tissues and paraneoplastic tissues to identify differentially expressed genes (DEGs). We conducted a Gene Set Enrichment Analysis and prognostic analysis by constructing competing endogenous RNA (ceRNA) networks; we performed a correlation analysis of the target genes and verified the targeting relationship of the ceRNA by cellular assays. We assessed the effects of the target genes on tumor cell proliferation, invasion and apoptosis by Cell Counting Kit-8 (CCK-8) assays, invasion assays, and caspase 3/7 assays, respectively. Results We identified 4,039 downregulated genes and 1,924 upregulated genes. The p53 pathway, cell-cycle pathway and mismatch-repair (MMR) pathway were activated, while the mitogen-activated protein kinase pathway was inhibited. Two ceRNA networks centered on the long non-coding RNAs (lncRNAs) MAGI2-AS3 and LINC01089 were constructed. MAGI2-AS3 was found to regulate five messenger RNAs (mRNAs) (i.e., MBNL2, ATP5L, FAM103A1, MDH1, and STXBP1) through three microRNAs (miRNAs), whereas LINC01089 was found to regulate six mRNAs (i.e., ZFP36L2, APBB2, PDLIM3, MYADM, PHF5A, and SLC26A9) through two miRNAs. The expression of these lncRNAs and mRNAs was significantly associated with prognosis (P<0.05). A significant correlation was also found between the expression of MAGI2-AS3 and MBNL2 (R=0.51), and both signatures were also significantly associated with prognosis. We also found that MAGI2-AS3 and MBNL2 had a regulatory relationship at the cellular level, for example, high expression of MBNL2 was noted to inhibit cancer cell proliferation and migration yet promote apoptosis. Conclusions MAGI-AS3 and MBNL2 are both differentially expressed in squamous cell carcinoma of the lung and are potential prognostic markers. A significant association was also found between MAGI2-AS3 and the expression of MBNL2 (R=0.51). High expression of MBNL2 inhibits cancer cell proliferation and migration, yet promotes cancer cell apoptosis.
Collapse
Affiliation(s)
- Xiaoxi Wan
- Department of Oncology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Chuanxia Zhang
- Department of Oncology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Mengyuan Kang
- Department of Oncology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Antonio Rossi
- Oncology Centre of Excellence, Therapeutic Science & Strategy Unit, IQVIA, Milan, Italy
| | - Taichiro Goto
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Yamanashi, Japan
| | - Nagarashee Seetharamu
- Division of Medical Oncology and Hematology, Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA
| | - Nobuhiko Seki
- Division of Medical Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Heng Lu
- Department of Oncology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Yang Zhang
- Department of Oncology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| |
Collapse
|
11
|
Szmajda-Krygier D, Nocoń Z, Pietrzak J, Krygier A, Balcerczak E. Assessment of Methylation in Selected ADAMTS Family Genes in Non-Small-Cell Lung Cancer. Int J Mol Sci 2025; 26:934. [PMID: 39940703 PMCID: PMC11816904 DOI: 10.3390/ijms26030934] [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: 12/20/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Alterations in the methylation of genetic material can influence carcinogenesis by the downregulation or overexpression of ADAMTS (a disintegrin-like and metalloprotease with thrombospondin motifs) protease genes. Through their proteolytic activity, these enzymes are also capable of promoting angiogenesis. Consequently, ADAMTS proteases can either facilitate or inhibit cancer progression. This study aimed to evaluate the methylation levels of the ADAMTS6, ADAMTS9, and ADAMTS12 genes in non-small-cell lung cancer (NSCLC) using data from bioinformatics databases. The focus was on differences between lung adenocarcinoma (LUAD) and lung squamous-cell carcinoma (LUSC) subtypes and their impact on patient overall survival (OS). ADAMTS6 gene expression is significantly reduced in LUSC, and analysis of ADAMTS9 gene expression showed a significantly reduced gene transcript level in LUAD and LUSC, while both NSCLC subtypes demonstrated ADAMTS12 upregulation. In LUSC, significantly elevated promoter methylation was found in all of the aforementioned genes, while in LUAD, higher promoter methylation was observed only for ADAMTS9 and ADAMTS12. The differential methylation region (DMR) pattern demonstrated by ADAMTS6, ADAMTS9, and ADAMTS12 is a useful tool for distinguishing normal from cancer cells. The areas under the curve (AUCs) ranged from 0.86 to 0.99 for both LUAD and LUSC subtypes. The methylation level of different CpG sites among selected ADAMTS members is related to patient survival, suggesting it may have value as a prognostic marker. The methylation degree of promoter regions in genes encoding ADAMTS family proteins could significantly influence LUSC and LUAD. Increased promoter methylation could also reduce certain gene expression, contributing to cancer progression. The expression levels and specific DMRs of ADAMTS genes may serve as prognostic markers correlating with patient OS. Assessing ADAMTS gene methylation could become a diagnostic tool for differentiating NSCLC subtypes and potentially guide therapeutic strategies. Further research is needed to fully understand the activity and mechanisms of ADAMTS family proteins.
Collapse
Affiliation(s)
- Dagmara Szmajda-Krygier
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Zuzanna Nocoń
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Jacek Pietrzak
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Adrian Krygier
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Ewa Balcerczak
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| |
Collapse
|
12
|
Zhang C, Yang C, Shi Q. Effects of Tp53 Gene Mutations on the Survival of Non-Small Cell Lung Cancer (NSCLC); A Short Review. Cancer Manag Res 2025; 17:65-82. [PMID: 39830995 PMCID: PMC11742633 DOI: 10.2147/cmar.s495006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. Mutations within the TP53 gene represent critical molecular events in NSCLC, contributing to the tumorigenesis in the pulmonary epithelial tissues. TP53 is a widely researched prognostic indicator in NSCLC, and pathological investigations have revealed a weak to mild negative predictive effect for TP53. Mutated p53 protein may have some pro-oncogenic impact, and the variations may change tumor inhibitors into oncogenes. The diverse mutational spectrum of TP53 in NSCLC with different mutations is linked to varied treatment responses. In contrast, first-line chemotherapeutics to this progress are limited, however, randomized trials with new chemotherapeutics have shown significant survival benefits. This review highlighted the critical influence of TP53 gene mutations on pathological-sensitivity and overall survival outcomes in NSCLC. Further research is needed to explore TP53 mutation-specific pathways and their effects on NSCLC progression and treatment effectiveness.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Oncology, Anhui Chest Hospital, Hefei, 230022, People’s Republic of China
- Anhui Medical University Clinical College of Chest, Hefei, 230022, People’s Republic of China
| | - Chao Yang
- Department of Urology, Anhui Provincial Children’s Hospital, Hefei, 230022, People’s Republic of China
| | - Qingming Shi
- Department of Oncology, Anhui Chest Hospital, Hefei, 230022, People’s Republic of China
- Anhui Medical University Clinical College of Chest, Hefei, 230022, People’s Republic of China
| |
Collapse
|
13
|
Chakraborty R, Dutta A, Mukhopadhyay R. TP53 mutations and MDM2 polymorphisms in breast and ovarian cancers: amelioration by drugs and natural compounds. Clin Transl Oncol 2025:10.1007/s12094-024-03841-6. [PMID: 39797946 DOI: 10.1007/s12094-024-03841-6] [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: 10/05/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025]
Abstract
Globally, breast and ovarian cancers are major health concerns in women and account for significantly high cancer-related mortality rates. Dysregulations and mutations in genes like TP53, BRCA1/2, KRAS and PTEN increase susceptibility towards cancer. Here, we discuss the impact of mutations in the key regulatory gene, TP53 and polymorphisms in its negative regulator MDM2 which are reported to accelerate cancer progression. Missense mutations, null mutations, transversions, transitions, and point mutations occurring in the TP53 gene can cause an increase in metastatic activity. This review discusses mutations occurring in exon regions of TP53, polymorphisms in MDM2 and their interaction with large ribosomal subunit protein (RPL) leading to cancer development. We also highlight the potential of small molecules e.g. p53 activators like XI-011, Tenovin-1, and Nutlin-3a for the treatment of breast and ovarian cancers. The therapeutic efficacy of natural compounds in amelioration of these two types of cancers is also discussed.
Collapse
Affiliation(s)
- Rituraj Chakraborty
- Inflammation and Cancer Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| | - Anupam Dutta
- Inflammation and Cancer Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| | - Rupak Mukhopadhyay
- Inflammation and Cancer Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India.
| |
Collapse
|
14
|
Izadi N, Solár P, Hašanová K, Zamani A, Akbar MS, Mrázová K, Bartošík M, Kazda T, Hrstka R, Joukal M. Breaking boundaries: role of the brain barriers in metastatic process. Fluids Barriers CNS 2025; 22:3. [PMID: 39780275 PMCID: PMC11708195 DOI: 10.1186/s12987-025-00618-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Brain metastases (BMs) are the most common intracranial tumors in adults and occur 3-10 times more frequently than primary brain tumors. Despite intensive multimodal therapies, including resection, radiotherapy, and chemotherapy, BMs are associated with poor prognosis and remain challenging to treat. BMs predominantly originate from primary lung (20-56%), breast (5-20%), and melanoma (7-16%) tumors, although they can arise from other cancer types less frequently. The metastatic cascade is a multistep process involving local invasion, intravasation into the bloodstream or lymphatic system, extravasation into normal tissue, and colonization of the distal site. After reaching the brain, circulating tumor cells (CTCs) breach the blood-brain barrier (BBB).The selective permeability of the BBB poses a significant challenge for therapeutic compounds, limiting the treatment efficacy of BMs. Understanding the mechanisms of tumor cell interactions with the BBB is crucial for the development of effective treatments. This review provides an in-depth analysis of the brain barriers, including the BBB, blood-spinal cord barrier, blood-meningeal barrier, blood-arachnoid barrier, and blood-cerebrospinal fluid barrier. It explores the molecular and cellular components of these barriers and their roles in brain metastasis, highlighting the importance of this knowledge for identifying druggable targets to prevent or limit BM formation.
Collapse
Affiliation(s)
- Nasim Izadi
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, St Anne University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Klaudia Hašanová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Maryam Shahidian Akbar
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Klára Mrázová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Martin Bartošík
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Tomáš Kazda
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Roman Hrstka
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czech Republic.
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
15
|
Mehta A, Mitra AG, Mani S, Dewan H, Mattoo S, Batra U. Prognosis of TP53 and Its Concomitant EGFR Mutation in Lung Cancer Especially Non-Small Cell Lung Cancer. Asian Pac J Cancer Prev 2025; 26:17-22. [PMID: 39873981 PMCID: PMC12082419 DOI: 10.31557/apjcp.2025.26.1.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Human Lung Carcinoma (LC) is among the most diagnosed cancers across the world among those non-small cell lung cancer (NSCLC) comprises about 85%. Next Generation Sequencing based detection of mutations are now well established in molecular oncology. With the advent of modern diagnostic methods, it is now well known that there are several mutations and gene rearrangements which are associated with the development of LC. Among those mutations, TP53 is the most prevalent with the concomitant EGFR mutation. METHODS In this retrospective study, a total number of 414 patients have been incorporated who have attended RGCIRC in the period between November 2015 to March 2024. Clinical stage has been determined as per NCCN Guideline version 2.2024. Nucleic Acid (DNA and RNA) from FFPE samples were extracted and detection of mutation was performed by Next generation sequencing (NGS) method. RESULTS All 414 patients opted for the customised NGS panel for lung cancer among those 203 patients were TP53 mutation and 87 patients were EGFR mutation positive. 62 patients were TP53-EGFR double mutation positive. The results of this study have shown that TP53 mutated patients show poor prognosis with conventional therapy. However, TP53-EGFR co-mutated patient's recovery rates are comparatively promising due to the availability of the targeted therapy of EGFR. CONCLUSION Studies have shown that TP53 mutation is unlikely to derive clinical benefit in LC patients and shows poorer prognosis when compared to TP53 wild type and EGFR mutated patients show improved recovery due to availability of the Kinase Inhibitor (KI) treatment. In this study we have observed and concluded that TP53-EGFR co-mutated group also shows promising prognosis for the application of KI treatment. A further large cohort study will establish this clinical observation and enlighten more therapeutically relevant information.
Collapse
Affiliation(s)
- Anurag Mehta
- Department of Research, RGCI&RC, Delhi, India.
- Molecular Lab, Department of Pathology, RGCI&RC, Delhi, India.
| | | | - Samson Mani
- Department of Research, RGCI&RC, Delhi, India.
| | - Himanshi Dewan
- Molecular Lab, Department of Pathology, RGCI&RC, Delhi, India.
| | - Sakshi Mattoo
- Molecular Lab, Department of Pathology, RGCI&RC, Delhi, India.
| | - Ullas Batra
- Department of Medical Oncology, RGCI&RC, Delhi, India.
| |
Collapse
|
16
|
Lee YS, Kwon RJ, Lee HS, Chung JH, Kim YS, Jeong HS, Park SJ, Lee SY, Kim T, Yoon SH. The Role of Pentacyclic Triterpenoids in Non-Small Cell Lung Cancer: The Mechanisms of Action and Therapeutic Potential. Pharmaceutics 2024; 17:22. [PMID: 39861671 PMCID: PMC11768946 DOI: 10.3390/pharmaceutics17010022] [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: 11/30/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Lung cancer remains a major global health problem because of its high cancer-related mortality rate despite advances in therapeutic approaches. Non-small cell lung cancer (NSCLC), a major subtype of lung cancer, is more amenable to surgical intervention in its early stages. However, the prognosis for advanced NSCLC remains poor, owing to limited treatment options. This underscores the growing need for novel therapeutic strategies to complement existing treatments and improve patient outcomes. In recent years, pentacyclic triterpenoids, a group of natural compounds, have emerged as promising candidates for cancer therapy due to their anticancer properties. Pentacyclic triterpenoids, such as lupeol, betulinic acid, betulin, oleanolic acid, ursolic acid, glycyrrhetinic acid, glycyrrhizin, and asiatic acid, have demonstrated the ability to inhibit cell proliferation and angiogenesis, induce apoptosis, suppress metastasis, and modulate inflammatory and immune pathways in NSCLC cell line models. These compounds exert their effects by modulating important signaling pathways such as NF-κB, PI3K/Akt, and MAPK. Furthermore, advances in drug delivery technologies such as nanocarriers and targeted delivery systems have improved the bioavailability and therapeutic efficacy of triterpenoids. However, despite promising preclinical data, rigorous clinical trials are needed to verify their safety and efficacy. This review explores the role of triterpenoids in NSCLC and therapeutic potential in preclinical models, focusing on their molecular mechanisms of action.
Collapse
Affiliation(s)
- Young-Shin Lee
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (Y.-S.L.); (R.J.K.); (H.S.L.)
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (Y.-S.L.); (R.J.K.); (H.S.L.)
| | - Hye Sun Lee
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (Y.-S.L.); (R.J.K.); (H.S.L.)
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA;
| | - Yun Seong Kim
- Division of Pulmonology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Han-Sol Jeong
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (H.-S.J.); (S.-J.P.); (S.Y.L.)
| | - Su-Jung Park
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (H.-S.J.); (S.-J.P.); (S.Y.L.)
| | - Seung Yeon Lee
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (H.-S.J.); (S.-J.P.); (S.Y.L.)
| | - Taehwa Kim
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea;
| | - Seong Hoon Yoon
- Division of Pulmonology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| |
Collapse
|
17
|
Xiang H, Kasajima R, Azuma K, Tagami T, Hagiwara A, Nakahara Y, Saito H, Igarashi Y, Wei F, Ban T, Yoshihara M, Nakamura Y, Sato S, Koizume S, Tamura T, Sasada T, Miyagi Y. Multi-omics analysis-based clinical and functional significance of a novel prognostic and immunotherapeutic gene signature derived from amino acid metabolism pathways in lung adenocarcinoma. Front Immunol 2024; 15:1361992. [PMID: 39735553 PMCID: PMC11671776 DOI: 10.3389/fimmu.2024.1361992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 07/30/2024] [Indexed: 12/31/2024] Open
Abstract
Background Studies have shown that tumor cell amino acid metabolism is closely associated with lung adenocarcinoma (LUAD) development and progression. However, the comprehensive multi-omics features and clinical impact of the expression of genes associated with amino acid metabolism in the LUAD tumor microenvironment (TME) are yet to be fully understood. Methods LUAD patients from The Cancer Genome Atlas (TCGA) database were enrolled in the training cohort. Using least absolute shrinkage and selection operator Cox regression analysis, we developed PTAAMG-Sig, a signature based on the expression of tumor-specific amino acid metabolism genes associated with overall survival (OS) prognosis. We evaluated its predictive performance for OS and thoroughly explored the effects of the PTAAMG-Sig risk score on the TME. The risk score was validated in two Gene Expression Omnibus (GEO) cohorts and further investigated against an original cohort of chemotherapy combined with immune checkpoint inhibitors (ICIs). Somatic mutation, chemotherapy response, immunotherapy response, gene set variation, gene set enrichment, immune infiltration, and plasma-free amino acids (PFAAs) profile analyses were performed to identify the underlying multi-omics features. Results TCGA datasets based PTAAMG-Sig model consisting of nine genes, KYNU, PSPH, PPAT, MIF, GCLC, ACAD8, TYRP1, ALDH2, and HDC, could effectively stratify the OS in LUAD patients. The two other GEO-independent datasets validated the robust predictive power of PTAAMG-Sig. Our differential analysis of somatic mutations in the high- and low-risk groups in TCGA cohort showed that the TP53 mutation rate was significantly higher in the high-risk group and negatively correlated with OS. Prediction from transcriptome data raised the possibility that PTAAMG-Sig could predict the response to chemotherapy and ICIs therapy. Our immunotherapy cohort confirmed the predictive ability of PTAAMG-Sig in the clinical response to ICIs therapy, which correlated with the infiltration of immune cells (e.g., T lymphocytes and nature killer cells). Corresponding to the concentrations of PFAAs, we discovered that the high PTAAMG-Sig risk score patients showed a significantly lower concentration of plasma-free α-aminobutyric acid. Conclusion In patients with LUAD, the PTAAMG-Sig effectively predicted OS, drug sensitivity, and immunotherapy outcomes. These findings are expected to provide new targets and strategies for personalized treatment of LUAD patients.
Collapse
Affiliation(s)
- Huihui Xiang
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Rika Kasajima
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Center for Cancer Genome Medicine, Kanagawa Cancer Center, Yokohama, Japan
| | - Koichi Azuma
- Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomoyuki Tagami
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kanagawa, Japan
| | - Asami Hagiwara
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kanagawa, Japan
| | - Yoshiro Nakahara
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Tatsuma Ban
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mitsuyo Yoshihara
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shinya Sato
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shiro Koizume
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
18
|
Dong L, Li L, Zhu L, Xu F, Zhang R, Li Q, Zhu Y, Zeng Z, Ding K. Multiomics analysis of homologous recombination deficiency across cancer types. BIOMOLECULES & BIOMEDICINE 2024; 25:71-81. [PMID: 39073402 PMCID: PMC11647252 DOI: 10.17305/bb.2024.10448] [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: 03/07/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
There remains ongoing debate regarding the association of homologous recombination deficiency (HRD) with patient survival across various malignancies, highlighting the need for a comprehensive understanding of HRD's role in different cancer types. Based on data from databases, we conducted a multivariable omics analysis on HRD in 33 cancer types, focusing mainly on 23 cancers in which HRD was significantly associated with patient overall survival (OS) rates. This analysis included the mechanisms related to patient prognosis, gene expression, gene mutation, and signaling pathways. In this study, HRD was found to be significantly associated with patient prognosis, but its impact varied among different cancers. HRD was linked to different outcomes for patients with distinct tumor subtypes and was correlated with clinical features such as clinical stage and tumor grade. Driver gene mutations, including TP53, MUC4, KRAS, HRAS, FLG, ANK3, BRCA2, ATRX, FGFR3, NFE2L2, MAP3K1, PIK3CA, CIC, FUBP1, ALB, CTNNB1, and MED12, were associated with HRD across specific cancer types. We also analyzed differentially expressed genes (DEGs) and differentially methylated regions (DMRs) in relation to HRD levels in these cancers. Furthermore, we explored the correlation between HRD and signaling pathways, as well as immune cell infiltration. Overall, our findings contribute to a comprehensive understanding of HRD's multifaceted role in cancer.
Collapse
Affiliation(s)
- Lin Dong
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Lin Li
- Department of Pathology, Tongling People’s Hospital, Tongling, Anhui, China
| | - Linyan Zhu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Fei Xu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pathology, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Rumeng Zhang
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Qiushuang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yong Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhutian Zeng
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Keshuo Ding
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
19
|
Wang H, Ma J, Lu J, Wang Y, Zhang B, Zhang H, Peng H. TMB is associated with the prognosis of egfr-mutated non-small cell lung cancer in Xuanwei, China. Biomark Med 2024; 18:1123-1133. [PMID: 39633593 DOI: 10.1080/17520363.2024.2432306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
AIMS Study on the prognostic characteristics and biomarkers of lung cancer in Xuanwei Patients. MATERIALS AND METHODS A retrospective analysis was conducted on the genetic data of 261 NSCLC patients, as well as the prognostic data of 133 patients. The relationship between prognosis and EGFR mutations in the Xuanwei and non Xuanwei cohorts was compared and analyzed. RESULTS The superior progression-free survival (PFS) in Xuanwei patients was primarily observed in those with EGFR-mutated tumors (p < 0.05). Further analysis of accompanying mutations and TMB levels revealed an association between high TMB and a favorable prognosis in EGFR-mutated Xuanwei NSCLC. CONCLUSION Xuanwei lung cancer differs from non Xuanwei patients in terms of prognosis and tumor mutation burden, and further research should be conducted.
Collapse
Affiliation(s)
- Han Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, P.R. China
| | - Junrui Ma
- School of Nursing, Yunnan University of Traditional Chinese Medicines, Kunming City, Yunnan Province, P.R. China
| | - Jiagui Lu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, P.R. China
| | - Yang Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, P.R. China
| | - Binli Zhang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, P.R. China
| | - Hushan Zhang
- Key Laboratory of Northeastern Yunnan Natural Medicine, Zhaotong Healthy Vocational College, Zhaotong City, Yunnan Province, China
- Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, Yunnan, P.R. China
- The Medical Department, 3D Medicines Inc, Shanghai, P.R. China
| | - Hao Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, P.R. China
| |
Collapse
|
20
|
Maddah MM, Hedayatizadeh-Omran A, Moosazadeh M, Alizadeh-Navaei R. Evaluation of the Prognostic Role of TP53 Gene Mutations in Prostate Cancer Outcome: A Systematic Review and Meta-Analysis. Clin Genitourin Cancer 2024; 22:102226. [PMID: 39393313 DOI: 10.1016/j.clgc.2024.102226] [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: 08/04/2024] [Revised: 08/31/2024] [Accepted: 09/07/2024] [Indexed: 10/13/2024]
Abstract
INTRODUCTION Prostate cancer, 1 of the most common cancers in men, is influenced by age, genetics, race, and lifestyle. The TP53 gene, encoding the p53 protein crucial for cell cycle regulation and DNA repair, is frequently mutated in metastatic prostate cancers. These mutations impact prognosis and resistance to treatments, underscoring the role of genetic factors in disease progression and therapeutic challenges. METHODS Databases such as PubMed, Scopus, and ISI were searched using the keywords "prostate cancer," "P53," "TP53," "survival," and "prognosis," along with manual searches in other sources. Initial screening and selection of articles were conducted independently and blinded by 2 reviewers, focusing on titles abstracts, and full texts when necessary. The Newcastle-Ottawa Scale (NOS) was used for full-text evaluation. Data were analyzed using STATA 11, with heterogeneity assessed using the I² index. RESULTS Overall survival (OS) for prostate cancer patients with TP53 mutations was approximately 13% lower than for those without mutations at 1 year, 20% lower at 3 years, and 16% lower at 5 years. TP53 mutations were also associated with faster disease progression and a 15% reduction in progression-free survival (PFS) over 1 year. The hazard ratio (HR) for death in patients with TP53 mutations was 1.76, and for PFS, it was 1.62, indicating a 76% increased risk of death and a 62% increased risk of disease progression. CONCLUSION TP53 mutations are associated with shorter survival and faster disease progression in prostate cancer, underscoring the importance of precise evaluation and management of these mutations in treatment.
Collapse
Affiliation(s)
- Mohammad Moein Maddah
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Science, Sari, Iran
| | - Akbar Hedayatizadeh-Omran
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Science, Sari, Iran
| | - Mahmood Moosazadeh
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Science, Sari, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Science, Sari, Iran.
| |
Collapse
|
21
|
Nisar H, Brauny M, Labonté FM, Schmitz C, Konda B, Hellweg CE. DNA Damage and Inflammatory Response of p53 Null H358 Non-Small Cell Lung Cancer Cells to X-Ray Exposure Under Chronic Hypoxia. Int J Mol Sci 2024; 25:12590. [PMID: 39684302 DOI: 10.3390/ijms252312590] [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: 10/17/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Hypoxia-induced radioresistance limits therapeutic success in cancer. In addition, p53 mutations are widespread in tumors including non-small cell lung carcinomas (NSCLCs), and they might modify the radiation response of hypoxic tumor cells. We therefore analyzed the DNA damage and inflammatory response in chronically hypoxic (1% O2, 48 h) p53 null H358 NSCLC cells after X-ray exposure. We used the colony-forming ability assay to determine cell survival, γH2AX immunofluorescence microscopy to quantify DNA double-strand breaks (DSBs), flow cytometry of DAPI-stained cells to measure cell cycle distribution, ELISAs to quantify IL-6 and IL-8 secretion in cell culture supernatants, and RNA sequencing to determine gene expression. Chronic hypoxia increased the colony-forming ability and radioresistance of H358 cells. It did not affect the formation or resolution of X-ray-induced DSBs. It reduced the fraction of cells undergoing G2 arrest after X-ray exposure and delayed the onset of G2 arrest. Hypoxia led to an earlier enhancement in cytokines secretion rate after X-irradiation compared to normoxic controls. Gene expression changes were most pronounced after the combined exposure to hypoxia and X-rays and pertained to senescence and different cell death pathways. In conclusion, hypoxia-induced radioresistance is present despite the absence of functional p53. This resistance is related to differences in clonogenicity, cell cycle regulation, cytokine secretion, and gene expression under chronic hypoxia, but not to differences in DNA DSB repair kinetics.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad 44000, Pakistan
| | - Melanie Brauny
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Faculty of Science & Faculty of Medicine, University of Tübingen, 72074 Tübingen, Germany
| | - Frederik M Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Department of Biology, Faculty of Mathematics and Natural Sciences, University of Cologne, 50923 Cologne, Germany
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
| | - Christine E Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
| |
Collapse
|
22
|
Li R, Liu X, Xu Y, Zhao J, Zhong W, Gao X, Chen M, Wang M. Remarkable pathological response to neoadjuvant tepotinib in lung adenocarcinoma with MET exon 14 skipping mutation: A case report. Thorac Cancer 2024; 15:2339-2343. [PMID: 39343987 PMCID: PMC11554545 DOI: 10.1111/1759-7714.15459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Mesenchymal-epithelial transition (MET) exon 14 (METex14) skipping mutation is a rare (3%-4%) driver mutation in non-small cell lung cancer (NSCLC). Tepotinib, a selective MET inhibitor, has shown promise in treating METex14 skipping-mutated NSCLC. However, its feasibility for perioperative application remains unclear. This report describes a 60-year-old man with stage IIIA (cT2N2M0) lung adenocarcinoma harboring a METex14 skipping mutation. After initial treatment with savolitinib was discontinued due to grade 4 transaminitis, the patient was switched to tepotinib, resulting in significant tumor regression. Six months later, further shrinkage was observed, and surgery revealed remarkable pathological response with no residual tumor in lymph nodes (ypT2N0M0, IB). Postoperative tepotinib continued, with no relapse at 6-month follow-up. This case highlights the potential of tepotinib as neoadjuvant therapy for resectable METex14 skipping-mutated NSCLC, warranting further clinical trials.
Collapse
Affiliation(s)
- Rongzhen Li
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yan Xu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jing Zhao
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wei Zhong
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoxing Gao
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Minjiang Chen
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mengzhao Wang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
23
|
Song Y, Zhou D, Zhang P, Zhu N, Guo R, Wang T, Zhuang F, Sun D. Heparanase accelerates the angiogenesis and inhibits the ferroptosis of p53-mutant non-small cell cancers in VEGF-dependent manner. Cytotechnology 2024; 76:503-517. [PMID: 39188651 PMCID: PMC11344742 DOI: 10.1007/s10616-024-00632-3] [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: 11/09/2023] [Accepted: 04/17/2024] [Indexed: 08/28/2024] Open
Abstract
The aim of this study is to explore the effects and specific mechanisms of heparanase on angiogenesis and iron deficiency anemia in TP53 mutant cancer. For this purpose, we conducted in vitro cell experiments and in vivo animal experiments respectively. In this study, we first analyzed the differential expression of heparanase in TP53 wild-type and mutant cells, and analyzed its effects on iron removal and angiogenesis in two types of CALU-1 and NCI-H358 cells. Secondly, we validated whether the mechanism of action of heparanase on TP53 mutant cells for iron removal and angiogenesis is related to VEGF. We applied the iron removal agonist erastin and VEGF inhibitor bevacizumab in both in vitro and in vivo experiments to validate the relationship between heparanase and VEGF in the mechanisms of iron removal and angiogenesis. The experimental results show that heparanase is highly expressed in TP53 mutated cancer cells, and has anti-ferroptosis and pro-angiogenic effects. Our experiment also confirmed that the effect of heparanase on TP53 mutant cancer's iron removal and angiogenesis is related to VEGF. In short, heparanase is highly expressed in p53 mutated lung cancer, and the mechanism of ferroptosis tolerance to TP53 mutated cancer is related to VEGF.
Collapse
Affiliation(s)
- Yaobo Song
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Dongmei Zhou
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Ping Zhang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Na Zhu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Ruijuan Guo
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Tian Wang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Feifei Zhuang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Dengjun Sun
- Department of Medical Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000 Shandong Province China
| |
Collapse
|
24
|
Zhang H, Liu S, Li B, Zhou X. IPFMC: an iterative pathway fusion approach for enhanced multi-omics clustering in cancer research. Brief Bioinform 2024; 25:bbae541. [PMID: 39470306 PMCID: PMC11514061 DOI: 10.1093/bib/bbae541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/13/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Using multi-omics data for clustering (cancer subtyping) is crucial for precision medicine research. Despite numerous methods having been proposed, current approaches either do not perform satisfactorily or lack biological interpretability, limiting the practical application of these methods. Based on the biological hypothesis that patients with the same subtype may exhibit similar dysregulated pathways, we developed an Iterative Pathway Fusion approach for enhanced Multi-omics Clustering (IPFMC), a novel multi-omics clustering method involving two data fusion stages. In the first stage, omics data are partitioned at each layer using pathway information, with crucial pathways iteratively selected to represent samples. Ultimately, the representation information from multiple pathways is integrated. In the second stage, similarity network fusion was applied to integrate the representation information from multiple omics. Comparative experiments with nine cancer datasets from The Cancer Genome Atlas (TCGA), involving systematic comparisons with 10 representative methods, reveal that IPFMC outperforms these methods. Additionally, the biological pathways and genes identified by our approach hold biological significance, affirming not only its excellent clustering performance but also its biological interpretability.
Collapse
Affiliation(s)
- Haoyang Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, People’s Republic of China
| | - Sha Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, People’s Republic of China
| | - Bingxin Li
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, People’s Republic of China
| | - Xionghui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, People’s Republic of China
- Key Laboratory of Smart Farming for Agricultural Animals, Ministry of Agriculture and Rural Affairs, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, People’s Republic of China
| |
Collapse
|
25
|
Ooi VY, Yeh TY. Recent Advances and Mechanisms of Phage-Based Therapies in Cancer Treatment. Int J Mol Sci 2024; 25:9938. [PMID: 39337427 PMCID: PMC11432602 DOI: 10.3390/ijms25189938] [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: 08/01/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
The increasing interest in bacteriophage technology has prompted its novel applications to treat different medical conditions, most interestingly cancer. Due to their high specificity, manipulability, nontoxicity, and nanosize nature, phages are promising carriers in targeted therapy and cancer immunotherapy. This approach is particularly timely, as current challenges in cancer research include damage to healthy cells, inefficiency in targeting, obstruction by biological barriers, and drug resistance. Some cancers are being kept at the forefront of phage research, such as colorectal cancer and HCC, while others like lymphoma, cervical cancer, and myeloma have not been retouched in a decade. Common mechanisms are immunogenic antigen display on phage coats and the use of phage as transporters to carry drugs, genes, and other molecules. To date, popular phage treatments being tested are gene therapy and phage-based vaccines using M13 and λ phage, with some vaccines having advanced to human clinical trials. The results from most of these studies have been promising, but limitations in phage-based therapies such as reticuloendothelial system clearance or diffusion inefficiency must be addressed. Before phage-based therapies for cancer can be successfully used in oncology practice, more in-depth research and support from local governments are required.
Collapse
Affiliation(s)
| | - Ting-Yu Yeh
- Agricultural Biotechnology Laboratory, Auxergen Inc., Riti Rossi Colwell Center, 701 E Pratt Street, Baltimore, MD 21202, USA
| |
Collapse
|
26
|
Nishitsuji K, Mito R, Ikezaki M, Yano H, Fujiwara Y, Matsubara E, Nishikawa T, Ihara Y, Uchimura K, Iwahashi N, Sakagami T, Suzuki M, Komohara Y. Impacts of cytoplasmic p53 aggregates on the prognosis and the transcriptome in lung squamous cell carcinoma. Cancer Sci 2024; 115:2947-2960. [PMID: 39031627 PMCID: PMC11462941 DOI: 10.1111/cas.16252] [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: 10/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/22/2024] Open
Abstract
The tumor suppressor TP53 gene, the most frequently mutated gene in human cancers, produces the product tumor protein p53, which plays an essential role in DNA damage. p53 protein mutations may contribute to tumorigenesis by loss of tumor suppressive functions and malignancy of cancer cells via gain-of-oncogenic functions. We previously reported that mutant p53 proteins form aggregates and that cytoplasmic p53 aggregates were associated with poor prognosis in human ovarian cancer. However, the prognostic impact of p53 aggregation in other tumors including lung squamous cell carcinoma (SCC) is poorly understood. Here, we demonstrated that lung SCC cases with cytoplasmic p53 aggregates had a significantly poor clinical prognosis. Analysis via patient-derived tumor organoids (PDOs) established from lung SCC patients and possessing cytoplasmic p53 aggregates showed that eliminating cytoplasmic p53 aggregates suppressed cell proliferation. RNA sequencing and transcriptome analysis of p53 aggregate-harboring PDOs indicated multiple candidate pathways involved in p53 aggregate oncogenic functions. With lung SCC-derived cell lines, we found that cytoplasmic p53 aggregates contributed to cisplatin resistance. This study thus shows that p53 aggregates are a predictor of poor prognosis in lung SCC and suggests that detecting p53 aggregates via p53 conventional immunohistochemical analysis may aid patient selection for platinum-based therapy.
Collapse
Affiliation(s)
- Kazuchika Nishitsuji
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
- Unité de Glycobiologie Structurale et FonctionnelleUMR 8576 CNRS, Université de LilleVilleneuve d'AscqFrance
| | - Remi Mito
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Respiratory Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Midori Ikezaki
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Eri Matsubara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Thoracic and Breast Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Taro Nishikawa
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Yoshito Ihara
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kenji Uchimura
- Unité de Glycobiologie Structurale et FonctionnelleUMR 8576 CNRS, Université de LilleVilleneuve d'AscqFrance
| | - Naoyuki Iwahashi
- Department of Obstetrics and Gynecology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Makoto Suzuki
- Department of Thoracic and Breast Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy AgingKumamoto UniversityKumamotoJapan
| |
Collapse
|
27
|
Zhang JN, Yang BB, Li LW, Xu H, Wang B, Yi ZL, Zhou XR, Liu H. Multi-omics pan-cancer analysis reveals the prognostic values and immunological functions of PPA2, with a spotlight on breast cancer. Front Immunol 2024; 15:1435502. [PMID: 39176095 PMCID: PMC11338811 DOI: 10.3389/fimmu.2024.1435502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Background Recently, the role of inorganic pyrophosphatase 2 (PPA2) has been remaining merely superficial in many tumors. Hence, the aim was to analyze the potential functions of PPA2 in pan-cancer, focusing on its role in breast cancer. Methods A systematic pan-cancer analysis conducted primarily utilizing various open databases such as TCGA and GTEx. We explored the clinical value of PPA2 as well as various biological functions, including expression levels and subcellular localization, multi-dimensional immune-correlation analysis, co-expression networks, and gene heterogeneity. In addition, we not only verified the function of PPA2 through cell experiments but also analyzed PPA2 at the single-cell level and its drug sensitivity. Results PPA2 is abnormally expressed in various tumors, and it is mainly distributed in mitochondria. Furthermore, the indicators (OS, DSS, DFI, and PFI) of analysis hint that PPA2 exhibits significant prognostic value. At the same time, the genomic heterogeneity (including TMB, MSI, MATH, and NEO) of PPA2 in pan-cancer was analyzed. Across multiple tumors, the results showed a close correlation between PPA2 expression levels and different immune signatures (such as immune cell infiltration). All of these indicate that PPA2 could potentially be applied in the guidance of immunotherapy. We also have demonstrated that PPA2 promoted the process of breast cancer. Finally, some potential therapeutic agents (such as Fulvestrant) targeting the abnormal expression of PPA2 are revealed. Conclusion In conclusion, the results demonstrated the great value of PPA2 in pan-cancer research, as well as its potential as a therapeutic target for breast tumors.
Collapse
Affiliation(s)
- Jia-Ning Zhang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Bei-Bei Yang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Lin-Wei Li
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Hao Xu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Bin Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Zi-Lu Yi
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xi-Rui Zhou
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
28
|
Li J, Xiong S, He P, Liang P, Li C, Zhong R, Cai X, Xie Z, Liu J, Cheng B, Chen Z, Liang H, Lao S, Chen Z, Shi J, Li F, Feng Y, Huo Z, Deng H, Yu Z, Wang H, Zhan S, Xiang Y, Wang H, Zheng Y, Lin X, He J, Liang W. Spatial whole exome sequencing reveals the genetic features of highly-aggressive components in lung adenocarcinoma. Neoplasia 2024; 54:101013. [PMID: 38850835 PMCID: PMC11208950 DOI: 10.1016/j.neo.2024.101013] [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: 01/07/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/10/2024]
Abstract
In invasive lung adenocarcinoma (LUAD), patients with micropapillary (MIP) or solid (SOL) components had a significantly poorer prognosis than those with only lepidic (LEP), acinar (ACI) or papillary (PAP) components. It is interesting to explore the genetic features of different histologic subtypes, especially the highly aggressive components. Based on a cohort of 5,933 patients, this study observed that in different tumor size groups, LUAD with MIP/SOL components showed a different prevalence, and patients with ALK alteration or TP53 mutations had a higher probability of developing MIP/SOL components. To control individual differences, this research used spatial whole-exome sequencing (WES) via laser-capture microdissection of five patients harboring these five coexistent components and identified genetic features among different histologic components of the same tumor. In tracing the evolution of components, we found that titin (TTN) mutation might serve as a crucial intratumor potential driver for MIP/SOL components, which was validated by a cohort of 146 LUAD patients undergoing bulk WES. Functional analysis revealed that TTN mutations enriched the complement and coagulation cascades, which correlated with the pathway of cell adhesion, migration, and proliferation. Collectively, the histologic subtypes of invasive LUAD were genetically different, and certain trunk genotypes might synergize with branching TTN mutation to develop highly aggressive components.
Collapse
Affiliation(s)
- Jianfu Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Shan Xiong
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Ping He
- Department of pathology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Peng Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Ran Zhong
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Xiuyu Cai
- Department of General Internal Medicine, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Zhanhong Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou 510120, China
| | - Jun Liu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Bo Cheng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Zhuxing Chen
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Hengrui Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Shen Lao
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Zisheng Chen
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Jiang Shi
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Feng Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Yi Feng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Zhenyu Huo
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Hongsheng Deng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Ziwen Yu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Haixuan Wang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Shuting Zhan
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Yang Xiang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Huiting Wang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Yongmin Zheng
- Department of pathology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Xiaodong Lin
- Department of pathology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China; Southern Medical University, Guangzhou 510120, China.
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China.
| |
Collapse
|
29
|
Kim H, Lee JK, Kim HR, Hong YJ. Enhanced Lung Cancer Detection Using a Combined Ratio of Antigen-Autoantibody Immune Complexes against CYFRA 21-1 and p53. Cancers (Basel) 2024; 16:2661. [PMID: 39123389 PMCID: PMC11312164 DOI: 10.3390/cancers16152661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
The early detection of lung cancer (LC) improves patient outcomes, but current methods have limitations. Autoantibodies against tumor-associated antigens have potential as early biomarkers. This study evaluated the 9G testTM Cancer/Lung, measuring circulating complexes of two antigen-autoantibody immune complexes (AIC) against their respective free antigens (CYFRA 21-1 and p53) for LC diagnosis. We analyzed 100 LC patients and 119 healthy controls using the 9G testTM Cancer/Lung, quantifying the levels of AICs (CYFRA 21-1-Anti-CYFRA 21-1 autoantibody immune complex (CIC) and p53-Anti-p53 autoantibody immune complex (PIC)), free antigens (CYFRA 21-1 and p53), and ratios of AICs/antigens (LC index). The levels of the CICs and PICs were significantly elevated in LC compared to the controls (p < 0.0062 and p < 0.0026), while free antigens showed no significant difference. The CIC/CYFRA 21-1 and PIC/p53 ratios were also significantly higher in LC (all, p < 0.0001). The LC index, when combining both ratios, exhibited the best diagnostic performance with an area under the curve (AUC) of 0.945, exceeding individual CICs, PICs, and free antigens (AUCs ≤ 0.887). At a cut-off of 3.60, the LC index achieved 81% sensitivity and 95% specificity for LC diagnosis. It detected early-stage (Stage I-II) LC with 87.5% sensitivity, exceeding its performance in advanced stages (72.7%). The LC index showed no significant differences based on age, gender, smoking status (former, current, or never smoker), or pack years smoked. The LC index demonstrates promising potential for early LC diagnosis, exceeding conventional free antigen markers.
Collapse
Affiliation(s)
- Heyjin Kim
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Jin Kyung Lee
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Hye-Ryoun Kim
- Division of Pulmonology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| | - Young Jun Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| |
Collapse
|
30
|
Liu L, Xiong Y, Zheng Z, Huang L, Song J, Lin Q, Tang B, Wong KC. AutoCancer as an automated multimodal framework for early cancer detection. iScience 2024; 27:110183. [PMID: 38989460 PMCID: PMC11233972 DOI: 10.1016/j.isci.2024.110183] [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: 12/22/2023] [Revised: 03/21/2024] [Accepted: 06/01/2024] [Indexed: 07/12/2024] Open
Abstract
Current studies in early cancer detection based on liquid biopsy data often rely on off-the-shelf models and face challenges with heterogeneous data, as well as manually designed data preprocessing pipelines with different parameter settings. To address those challenges, we present AutoCancer, an automated, multimodal, and interpretable transformer-based framework. This framework integrates feature selection, neural architecture search, and hyperparameter optimization into a unified optimization problem with Bayesian optimization. Comprehensive experiments demonstrate that AutoCancer achieves accurate performance in specific cancer types and pan-cancer analysis, outperforming existing methods across three cohorts. We further demonstrated the interpretability of AutoCancer by identifying key gene mutations associated with non-small cell lung cancer to pinpoint crucial factors at different stages and subtypes. The robustness of AutoCancer, coupled with its strong interpretability, underscores its potential for clinical applications in early cancer detection.
Collapse
Affiliation(s)
- Linjing Liu
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Ying Xiong
- Department of Computer Science, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Zetian Zheng
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Lei Huang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Qiuzhen Lin
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Buzhou Tang
- Department of Computer Science, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| |
Collapse
|
31
|
Liu Y, Yalavarthi S, Yang F, Abdul-Rashid Y, Tang S, Long Z, Qin Y, Wu K, Wang Z. Insights into treatment-specific prognostic somatic mutations in NSCLC from the AACR NSCLC GENIE BPC cohort analysis. BMC Pulm Med 2024; 24:309. [PMID: 38956553 PMCID: PMC11218090 DOI: 10.1186/s12890-024-03124-4] [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: 03/15/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Treatment of non-small lung cancer (NSCLC) has evolved in recent years, benefiting from advances in immunotherapy and targeted therapy. However, limited biomarkers exist to assist clinicians and patients in selecting the most effective, personalized treatment strategies. Targeted next-generation sequencing-based genomic profiling has become routine in cancer treatment and generated crucial clinicogenomic data over the last decade. This has made the development of mutational biomarkers for drug response possible. METHODS To investigate the association between a patient's responses to a specific somatic mutation treatment, we analyzed the NSCLC GENIE BPC cohort, which includes 2,004 tumor samples from 1,846 patients. RESULTS We identified somatic mutation signatures associated with response to immunotherapy and chemotherapy, including carboplatin-, cisplatin-, pemetrexed- or docetaxel-based chemotherapy. The prediction power of the chemotherapy-associated signature was significantly affected by epidermal growth factor receptor (EGFR) mutation status. Therefore, we developed an EGFR wild-type-specific mutation signature for chemotherapy selection. CONCLUSION Our treatment-specific gene signatures will assist clinicians and patients in selecting from multiple treatment options.
Collapse
Affiliation(s)
- Yi Liu
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Sindhu Yalavarthi
- Department of Nanoscience, The Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro, Greensboro, NC, 27401, USA
| | - Fan Yang
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Yusif Abdul-Rashid
- Department of Nanoscience, The Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro, Greensboro, NC, 27401, USA
| | - Shenkun Tang
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Zihe Long
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Yongkai Qin
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Kerui Wu
- Department of Nanoscience, The Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro, Greensboro, NC, 27401, USA
| | - Zhifei Wang
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China.
| |
Collapse
|
32
|
Benitez DA, Cumplido-Laso G, Olivera-Gómez M, Del Valle-Del Pino N, Díaz-Pizarro A, Mulero-Navarro S, Román-García A, Carvajal-Gonzalez JM. p53 Genetics and Biology in Lung Carcinomas: Insights, Implications and Clinical Applications. Biomedicines 2024; 12:1453. [PMID: 39062026 PMCID: PMC11274425 DOI: 10.3390/biomedicines12071453] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The TP53 gene is renowned as a tumor suppressor, playing a pivotal role in overseeing the cell cycle, apoptosis, and maintaining genomic stability. Dysregulation of p53 often contributes to the initiation and progression of various cancers, including lung cancer (LC) subtypes. The review explores the intricate relationship between p53 and its role in the development and progression of LC. p53, a crucial tumor suppressor protein, exists in various isoforms, and understanding their distinct functions in LC is essential for advancing our knowledge of this deadly disease. This review aims to provide a comprehensive literature overview of p53, its relevance to LC, and potential clinical applications.
Collapse
Affiliation(s)
- Dixan A. Benitez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| | | | | | | | | | | | | | - Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| |
Collapse
|
33
|
Zhang X, Yu D, Tang P, Chen F. Insights into the role of mitophagy in lung cancer: current evidence and perspectives. Front Pharmacol 2024; 15:1420643. [PMID: 38962310 PMCID: PMC11220236 DOI: 10.3389/fphar.2024.1420643] [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: 04/20/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Lung cancer, recognized globally as a leading cause of malignancy-associated morbidity and mortality, is marked by its high prevalence and lethality, garnering extensive attention within the medical community. Mitophagy is a critical cellular process that plays a crucial role in regulating metabolism and ensuring quality control within cells. Its relevance to lung cancer has garnered significant attention among researchers and scientists. Mitophagy's involvement in lung cancer encompasses its initiation, progression, metastatic dissemination and treatment. The regulatory landscape of mitophagy is complex, involving numerous signaling proteins and pathways that may exhibit aberrant alterations or mutations within the tumor environment. In the field of treatment, the regulation of mitophagy is considered key to determining cancer chemotherapy, radiation therapy, other treatment options, and drug resistance. Contemporary investigations are directed towards harnessing mitophagy modulators, both inhibitors and activators, in therapeutic strategies, with an emphasis on achieving specificity to minimize collateral damage to healthy cellular populations. Furthermore, molecular constituents and pathways affiliated with mitophagy, serving as potential biomarkers, offer promising avenues for enhancing diagnostic accuracy, prognostic assessment, and prediction of therapeutic responses in lung cancer. Future endeavors will also involve investigating the impact of mitophagy on the composition and function of immune cells within the tumor microenvironment, aiming to enhance our understanding of how mitophagy modulates the immune response to lung cancer. This review aims to comprehensively overview recent advancements about the role of mitophagy in the tumor genesis, progenesis and metastasis, and the impact of mitophagy on the treatment of lung cancer. We also discussed the future research direction of mitophagy in the field of lung cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Dongzhi Yu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peng Tang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fengshou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
34
|
Dong X, Shao C, Xu S, Tu J, Xu W, Chen D, Tang Y. Construction and validation of a prognostic signature based on anoikis-related lncRNAs in lung adenocarcinoma. Aging (Albany NY) 2024; 16:9899-9917. [PMID: 38850527 PMCID: PMC11210241 DOI: 10.18632/aging.205905] [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: 08/11/2023] [Accepted: 05/02/2024] [Indexed: 06/10/2024]
Abstract
Lung adenocarcinoma (LUAD) is the most common type of lung cancer and is characterized by a high death rate and a poor prospect for survival. Anoikis, which is a kind of programmed cell apoptosis, is an important factor in the advancement of tumors. Nonetheless, the function of anoikis-related lncRNAs (ARLRs) in LUAD is still not well understood. The TCGA database was queried for genomic and clinical information. A prognostic signature for ARLRs was established via the use of coexpression analysis and Cox regression. Validation of the model's accuracy was conducted utilizing K-M curves and receiver operating characteristic (ROC) curves, and the signature was utilized to develop a nomogram. LncRNAs were implicated in the progression of tumors, as determined by functional enrichment analysis. There was an improvement in prognosis, increased immune cell infiltration, and higher immune scores among the low-risk patients. Additionally, we found that the two groups had varied anticancer drug sensitivities, which could help guide treatment. The impact of one ARLR, AC026355.2, on migration and invasion was validated by in vitro experiments in LUAD cells. Herein, a new lncRNA signature associated with anoikis was identified and estimated, potentially serving as a prognostic indicator for LUAD patients.
Collapse
Affiliation(s)
- Xiaoqi Dong
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Chuan Shao
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Shuguang Xu
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Jinjing Tu
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Wenjing Xu
- Ningbo University Health Science Center, Ningbo, China
| | - Dahua Chen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Yaodong Tang
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| |
Collapse
|
35
|
Boța M, Vlaia L, Jîjie AR, Marcovici I, Crişan F, Oancea C, Dehelean CA, Mateescu T, Moacă EA. Exploring Synergistic Interactions between Natural Compounds and Conventional Chemotherapeutic Drugs in Preclinical Models of Lung Cancer. Pharmaceuticals (Basel) 2024; 17:598. [PMID: 38794168 PMCID: PMC11123751 DOI: 10.3390/ph17050598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
In the current work, the synergy between natural compounds and conventional chemotherapeutic drugs is comprehensively reviewed in light of current preclinical research findings. The prognosis for lung cancer patients is poor, with a 5-year survival rate of 18.1%. The use of natural compounds in combination with conventional chemotherapeutic drugs has gained significant attention as a potential novel approach in the treatment of lung cancer. The present work highlights the importance of finding more effective therapies to increase survival rates. Chemotherapy is a primary treatment option for lung cancer but it has limitations such as reduced effectiveness because cancer cells become resistant. Natural compounds isolated from medicinal plants have shown promising anticancer or chemopreventive properties and their synergistic effect has been observed when combined with conventional therapies. The combined use of an anti-cancer drug and a natural compound exhibits synergistic effects, enhancing overall therapeutic actions against cancer cells. In conclusion, this work provides an overview of the latest preclinical research on medicinal plants and plant-derived compounds as alternative or complementary treatment options for lung cancer chemotherapy and discusses the potential of natural compounds in treating lung cancer with minimal side effects.
Collapse
Affiliation(s)
- Mihaela Boța
- Department II—Pharmaceutical Technology, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (M.B.); (L.V.)
| | - Lavinia Vlaia
- Department II—Pharmaceutical Technology, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (M.B.); (L.V.)
- Formulation and Technology of Drugs Research Center, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Alex-Robert Jîjie
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Iasmina Marcovici
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Flavia Crişan
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Cristian Oancea
- Discipline of Pneumology, Department of Infectious Diseases, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania;
| | - Cristina Adriana Dehelean
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Tudor Mateescu
- Department of Thoracic Surgery, Clinical Hospital for Infectious Diseases and Pneumophthiology Dr. Victor Babes, 13 Gheorghe Adam Street, RO-300310 Timisoara, Romania;
| | - Elena-Alina Moacă
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| |
Collapse
|
36
|
Chang F, Xi B, Chai X, Wang X, Ma M, Fan Y. Molecular mechanism of radiation tolerance in lung adenocarcinoma cells using single-cell RNA sequencing. J Cell Mol Med 2024; 28:e18378. [PMID: 38760895 PMCID: PMC11101670 DOI: 10.1111/jcmm.18378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/20/2024] Open
Abstract
The efficacy of radiotherapy, a cornerstone in the treatment of lung adenocarcinoma (LUAD), is profoundly undermined by radiotolerance. This resistance not only poses a significant clinical challenge but also compromises patient survival rates. Therefore, it is important to explore this mechanism for the treatment of LUAD. Multiple public databases were used for single-cell RNA sequencing (scRNA-seq) data. We filtered, normalized and downscaled scRNA-seq data based on the Seurat package to obtain different cell subpopulations. Subsequently, the ssGSEA algorithm was used to assess the enrichment scores of the different cell subpopulations, and thus screen the cell subpopulations that are most relevant to radiotherapy tolerance based on the Pearson method. Finally, pseudotime analysis was performed, and a preliminary exploration of gene mutations in different cell subpopulations was performed. We identified HIST1H1D+ A549 and PIF1+ A549 as the cell subpopulations related to radiotolerance. The expression levels of cell cycle-related genes and pathway enrichment scores of these two cell subpopulations increased gradually with the extension of radiation treatment time. Finally, we found that the proportion of TP53 mutations in patients who had received radiotherapy was significantly higher than that in patients who had not received radiotherapy. We identified two cellular subpopulations associated with radiotherapy tolerance, which may shed light on the molecular mechanisms of radiotherapy tolerance in LUAD and provide new clinical perspectives.
Collapse
Affiliation(s)
- Feiyun Chang
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Bozhou Xi
- The Second Clinical Medical SchoolShanxi Medical UniversityTaiyuanChina
| | - Xinchun Chai
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Xiuyan Wang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, Shenzhen YuceBioTechnology Co., LtdShenzhenChina
| | - Manyuan Ma
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, Shenzhen YuceBioTechnology Co., LtdShenzhenChina
| | - Yafeng Fan
- Department of Respiration, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
37
|
Frille A, Boeschen M, Wirtz H, Stiller M, Bläker H, von Laffert M. TP53 co-mutations in advanced lung adenocarcinoma: comparative bioinformatic analyses suggest ambivalent character on overall survival alongside KRAS, STK11 and KEAP1 mutations. Front Oncol 2024; 14:1357583. [PMID: 39156705 PMCID: PMC11327858 DOI: 10.3389/fonc.2024.1357583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/05/2024] [Indexed: 08/20/2024] Open
Abstract
Background Recently, we could show that the co-mutations of KRAS + KEAP1, STK11 + KEAP1 and KRAS + STK11 + KEAP1 lead to a significantly shorter median overall survival (mOS) across treatments by analyzing multiple datasets. TP53, a tumor suppressor gene, plays a crucial role in regulating cell cycle progression. Its mutations occur in approximately 40-50% of non-small lung cancer (NSCLC). Co-occurrence of all four mentioned mutations has been a matter of debate for years. The aim of this study was to assess the distribution of these four mutations and the influence of the different co-mutational patterns on survival. Methods We present a comparative bioinformatic analysis and refer to data of 4,109 patients with lung adenocarcinoma (LUAD). Results Most of the mutations within the LUAD belong to TP53-only (29.0%), quadruple-negative (25.9%) and KRAS-only (13.4%). Whereas TP53-mutations seem to have protective effects in the context of further KEAP1- and KRAS + KEAP1-alterations (improved mOS), their role seems contrary if acquired in an already existing combination of mutations as KRAS + STK11, KRAS + STK11 + KEAP1 and STK1 + KEAP1. TP53 co-mutationshad a negative influence on KRAS-only mutated LUAD (mOS reduced significantly by more than 30%). Discussion These data underline the need for complex mutational testing to estimate prognosis more accurately in patients with advanced LUAD.
Collapse
Affiliation(s)
- Armin Frille
- Department of Respiratory Medicine, Leipzig University, Leipzig, Germany
| | - Myriam Boeschen
- Institute of Pathology, Leipzig University, Leipzig, Germany
| | - Hubert Wirtz
- Department of Respiratory Medicine, Leipzig University, Leipzig, Germany
| | - Mathias Stiller
- Institute of Pathology, Leipzig University, Leipzig, Germany
| | - Hendrik Bläker
- Institute of Pathology, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
38
|
Wei J, Ji K, Zhang Y, Zhang J, Wu X, Ji X, Zhou K, Yang X, Lu H, Wang A, Bu Z. Exploration of molecular markers related to chemotherapy efficacy of hepatoid adenocarcinoma of the stomach. Cell Oncol (Dordr) 2024; 47:677-693. [PMID: 37943484 DOI: 10.1007/s13402-023-00892-9] [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] [Accepted: 10/08/2023] [Indexed: 11/10/2023] Open
Abstract
PURPOSE Preoperative neoadjuvant chemotherapy may not improve the prognosis of patients with hepatoid adenocarcinoma of the stomach (HAS), a rare pathological type of gastric cancer. Thus, the study aimed at the genomic and transcriptomic impacts of preoperative chemotherapy on HAS. METHODS Patients with HAS who underwent surgical resection at Peking University Cancer Hospital were retrospectively included in this study. Whole exome sequencing and transcriptome sequencing were performed on pre-chemotherapy, non-chemotherapy and post-chemotherapy samples. We then compared the alterations in molecular markers between the post-chemotherapy and non-chemotherapy groups, and between the chemotherapy-effective and chemotherapy-ineffective groups, respectively. RESULTS A total of 79 tumor samples from 72 patients were collected. Compared to the non-chemotherapy group, the mutation frequencies of several genes were changed after chemotherapy, including TP53. In addition, there was a significant increase in the frequency of frameshift mutations and cytosine transversion to adenine (C > A), appearance of COSMIC signature 6 and 14, and a reduced gene copy number amplification. Interestingly, the same phenomenon was observed in chemotherapy-ineffective patients. In addition, many HAS patients had ERBB2, FGFR2, MET and HGF gene amplification. Moreover, the expression of immune-related genes, especially those related to lymphocyte activation, was down-regulated after chemotherapy. CONCLUSION Chemotherapy is closely associated with changes in the molecular characteristics of HAS. After chemotherapy, at genomic and transcriptome level, many features were altered. These changes may be molecular markers of poor chemotherapeutic efficacy and play an important role in chemoresistance in HAS. In addition, ERBB2, FGFR2, MET and HGF gene amplification may be potential therapeutic targets for HAS.
Collapse
Affiliation(s)
- Jingtao Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Ke Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yue Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Ji Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xiaojiang Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xin Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Kai Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xuesong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Hongfeng Lu
- Berry Genomics Corporation, Beijing, 102206, China
| | - Anqiang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Zhaode Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
39
|
Prasad R, Sharma K, Bhutani K, Prasad S, Manhas S, Kishan J. Identification of Genetic Variants in Exon 4 of TP53 in Lung Carcinoma and in Silico Prediction of Their Significance. Indian J Clin Biochem 2024; 39:276-282. [PMID: 38577139 PMCID: PMC10987423 DOI: 10.1007/s12291-022-01099-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 11/09/2022] [Indexed: 12/13/2022]
Abstract
Lung cancer is a severe and the leading cause of cancer related deaths in men and women all over the world. Tumor suppressor protein (TP53) encoded by the TP53 gene which plays a pivotal role in various cellular tumor suppression processes viz cell cycle arrest and apoptosis. Henceforth, the present study was aimed to TP53 exon4 variants from lung carcinoma. Histopathologic and clinically proven 20 patients of lung cancer were enrolled in this study the average age of patients was 45 ± 8 years which categorized as early onset of lung cancer. Genomic DNA was isolated from the blood specimen of patients. Extracted DNA was subjected to PCR amplification for exon 4 of TP53 using appropriate primers and subsequently amplified products were applied to nucleotide alterations via using the DNA sanger sequencing. The genetic analysis documented five variants in exon4 of TP53 which include viz. 4 substitutions [c.215 > C at codon 72, C. 358-359AA > GG at codon 120] were highly prevalent, occurring in 63% and 25% frequency in patients. Other two variants viz. C. 358 A > C at codon 120, C. 365T > G at codon 122 were present at frequency of 15% whilst one deletion variant [152 del C] was found with 5% frequency. Furthermore, alterations on codon 72, 120,122 and 51 were characterized as possibly damaging by Poly Phen-2 and decreased stability using stability bioinformatic tool. Taken together all these findings infer that TP53 gene involved in modulation and susceptibility to lung cancer.
Collapse
Affiliation(s)
- Rajendra Prasad
- Department of Biochemistry, M.M. Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala India
| | - Kirti Sharma
- Department of Biochemistry, M.M. Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala India
| | - Karanpreet Bhutani
- Department of Biochemistry, M.M. Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala India
| | - Suvarna Prasad
- Department of Biochemistry, M.M. Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala India
- Department of Biochemistry, AIIMS, Deoghar, India
| | - Sunita Manhas
- Department of Biochemistry, M.M. Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala India
| | - Jai Kishan
- Department of Respiratory Medicine, M.M. Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala India
| |
Collapse
|
40
|
Lee JY, Bhandare RR, Boddu SHS, Shaik AB, Saktivel LP, Gupta G, Negi P, Barakat M, Singh SK, Dua K, Chellappan DK. Molecular mechanisms underlying the regulation of tumour suppressor genes in lung cancer. Biomed Pharmacother 2024; 173:116275. [PMID: 38394846 DOI: 10.1016/j.biopha.2024.116275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Tumour suppressor genes play a cardinal role in the development of a large array of human cancers, including lung cancer, which is one of the most frequently diagnosed cancers worldwide. Therefore, extensive studies have been committed to deciphering the underlying mechanisms of alterations of tumour suppressor genes in governing tumourigenesis, as well as resistance to cancer therapies. In spite of the encouraging clinical outcomes demonstrated by lung cancer patients on initial treatment, the subsequent unresponsiveness to first-line treatments manifested by virtually all the patients is inherently a contentious issue. In light of the aforementioned concerns, this review compiles the current knowledge on the molecular mechanisms of some of the tumour suppressor genes implicated in lung cancer that are either frequently mutated and/or are located on the chromosomal arms having high LOH rates (1p, 3p, 9p, 10q, 13q, and 17p). Our study identifies specific genomic loci prone to LOH, revealing a recurrent pattern in lung cancer cases. These loci, including 3p14.2 (FHIT), 9p21.3 (p16INK4a), 10q23 (PTEN), 17p13 (TP53), exhibit a higher susceptibility to LOH due to environmental factors such as exposure to DNA-damaging agents (carcinogens in cigarette smoke) and genetic factors such as chromosomal instability, genetic mutations, DNA replication errors, and genetic predisposition. Furthermore, this review summarizes the current treatment landscape and advancements for lung cancers, including the challenges and endeavours to overcome it. This review envisages inspired researchers to embark on a journey of discovery to add to the list of what was known in hopes of prompting the development of effective therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Sciences, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Richie R Bhandare
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates.
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates
| | - Afzal B Shaik
- St. Mary's College of Pharmacy, St. Mary's Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological University Kakinada, Chebrolu, Guntur, Andhra Pradesh 522212, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Lakshmana Prabu Saktivel
- Department of Pharmaceutical Technology, University College of Engineering (BIT Campus), Anna University, Tiruchirappalli 620024, India
| | - Gaurav Gupta
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University, PO Box 9, Solan, Himachal Pradesh 173229, India
| | - Muna Barakat
- Department of Clinical Pharmacy & Therapeutics, Applied Science Private University, Amman-11937, Jordan
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara 144411, India; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Kamal Dua
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
41
|
Najafiyan B, Bokaii Hosseini Z, Esmaelian S, Firuzpour F, Rahimipour Anaraki S, Kalantari L, Hheidari A, Mesgari H, Nabi-Afjadi M. Unveiling the potential effects of resveratrol in lung cancer treatment: Mechanisms and nanoparticle-based drug delivery strategies. Biomed Pharmacother 2024; 172:116207. [PMID: 38295754 DOI: 10.1016/j.biopha.2024.116207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 03/03/2024] Open
Abstract
Lung cancer ranks among the most prevalent forms of cancer and remains a significant factor in cancer-related mortality across the world. It poses significant challenges to healthcare systems and society as a whole due to its high incidence, mortality rates, and late-stage diagnosis. Resveratrol (RV), a natural compound found in various plants, has shown potential as a nanomedicine for lung cancer treatment. RV has varied effects on cancer cells, including promoting apoptosis by increasing pro-apoptotic proteins (Bax and Bak) and decreasing anti-apoptotic proteins (Bcl-2). It also hinders cell proliferation by influencing important signaling pathways (MAPK, mTOR, PI3K/Akt, and Wnt/β-catenin) that govern cancer progression. In addition, RV acts as a potent antioxidant, diminishing oxidative stress and safeguarding cells against DNA damage. However, using RV alone in cancer treatment has drawbacks, such as low bioavailability, lack of targeting ability, and susceptibility to degradation. In contrast, nanoparticle-based delivery systems address these limitations and hold promise for improving treatment outcomes in lung cancer; nanoparticle formulations of RV offer advantages such as improved drug delivery, increased stability, controlled release, and targeted delivery to lung cancer cells. This article will provide an overview of lung cancer, explore the potential of RV as a therapeutic agent, discuss the benefits and challenges of nanoparticle-based drug delivery, and highlight the promise of RV nanoparticles for cancer treatment, including lung cancer. By optimizing these systems for clinical application, future studies aim to enhance overall treatment outcomes and improve the prognosis for lung cancer patients.
Collapse
Affiliation(s)
- Behnam Najafiyan
- Faculty of Pharmacy, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Samar Esmaelian
- Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Faezeh Firuzpour
- Student of Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
42
|
Siwachat S, Tantraworasin A, Lertprasertsuke N, Saeteng S. Frequency and predictive factors of nodal micro-metastasis (NMM) in resectable non-small cell lung cancer. J Thorac Dis 2024; 16:1270-1278. [PMID: 38505058 PMCID: PMC10944788 DOI: 10.21037/jtd-23-1240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/12/2024] [Indexed: 03/21/2024]
Abstract
Background The frequency of lymph nodal micrometastasis (NMM) in resectable non-small cell lung cancer (NSCLC) is frequently underestimated when relying solely on standard hematoxylin and eosin staining during pathological examination. Methods This is a retrospective cross-sectional diagnostic research. Medical records of resectable pN0 NSCLC patients who underwent curative resection in Maharaj Nakorn Chiang Mai Hospital between January 2006 to December 2017 were retrospectively reviewed. Immunohistochemistry (IHC) staining using cytokeratin AE1/AE3, p53 and BerEP4 markers was employed to detect NMM. Primary objective of this study was to determine frequency of NMM in pN0 resectable NSCLC. Results This study included 98 patients with pN0 NSCLC, of which 47 were male and 51 were female. NMM was detected in 21 of 98 patients (21.43%). Lymph node station 10 and 7 were the most common site of micrometastasis among patients with N1 and N2 micrometastasis, respectively. Cytokeratin AE1/AE3 was the most sensitive antibody in detecting micrometastasis in lymph nodes, identifying 25 out of 27 positive lymph nodes. Tumor size greater than 4 cm was a statistically significant predictive factor for NMM with risk ratio 6.69 [95% confidence interval (CI): 2.38-18.85, P<0.001]. Conclusions NMM was identified in 21.43% of pN0 resectable NSCLC patients and tumor size greater than 4 cm is predictive factor for NMM.
Collapse
Affiliation(s)
- Sophon Siwachat
- General Thoracic Surgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
- Clinical Epidemiology and Clinical Statistic Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apichat Tantraworasin
- General Thoracic Surgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
- Clinical Epidemiology and Clinical Statistic Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nirush Lertprasertsuke
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Somcharoen Saeteng
- General Thoracic Surgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
43
|
Kanagasabai T, Dunbar Z, Ochoa SG, Farris T, Dhandayuthapani S, Wijeratne EMK, Gunatilaka AAL, Shanker A. Bortezomib in Combination with Physachenolide C Reduces the Tumorigenic Properties of KRAS mut/P53 mut Lung Cancer Cells by Inhibiting c-FLIP. Cancers (Basel) 2024; 16:670. [PMID: 38339421 PMCID: PMC10854725 DOI: 10.3390/cancers16030670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Defects in apoptosis regulation are one of the classical features of cancer cells, often associated with more aggressiveness and failure to therapeutic options. We investigated the combinatorial antitumor effects of a natural product, physachenolide C (PCC) and bortezomib, in KRASmut/P53mut lung cancer cells and xenograft mice models. METHODS The in vitro anticancer effects of the bortezomib and PCC combination were investigated using cell viability, migration, and invasion assays in 344SQ, H23, and H358 cell lines. Furthermore, the effects of combination treatment on the critical parameters of cellular metabolism, including extracellular acidification rate (ECAR) and mitochondrial oxidative phosphorylation based on the oxygen consumption rate of cancer cells were assessed using Seahorse assay. Finally, the antitumor effect of the bortezomib (1 mg/kg) and PCC (10 mg/kg) combination was evaluated using xenograft mice models. RESULTS Our data showed that the bortezomib-PCC combination was more effective in reducing the viability of lung cancer cells in comparison with the individual treatments. Similarly, the combination treatment showed a significant inhibition of cell migration and invasion of cancer cells. Additionally, the key anti-apoptotic protein c-FLIP was significantly inhibited along with a substantial reduction in the key parameters of cellular metabolism in cancer cells. Notably, the bortezomib or PCC inhibited the tumor growth compared to the control group, the tumor growth inhibition was much more effective when bortezomib was combined with PCC in tumor xenograft mice models. CONCLUSION These findings demonstrate that PCC sensitizes cancer cells to bortezomib, potentially improving the antitumor effects against KRASmut/P53mut lung cancer cells, with an enhanced efficacy of combination treatments without causing significant side effects.
Collapse
Affiliation(s)
- Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.K.); (T.F.)
| | - Zerick Dunbar
- Department of Microbiology, Immunology & Physiology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Salvador González Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Tonie Farris
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.K.); (T.F.)
| | | | - E. M. Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture, Life and Environmental Sciences, The University of Arizona, Tucson, AZ 85719, USA; (E.M.K.W.)
| | - A. A. Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture, Life and Environmental Sciences, The University of Arizona, Tucson, AZ 85719, USA; (E.M.K.W.)
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
44
|
Dwivedi K, Rajpal A, Rajpal S, Kumar V, Agarwal M, Kumar N. XL 1R-Net: Explainable AI-driven improved L 1-regularized deep neural architecture for NSCLC biomarker identification. Comput Biol Chem 2024; 108:107990. [PMID: 38000327 DOI: 10.1016/j.compbiolchem.2023.107990] [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: 05/31/2023] [Revised: 10/29/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND AND OBJECTIVE Non-small cell lung cancer (NSCLC) exhibits intrinsic molecular heterogeneity, primarily driven by the mutation of specific biomarkers. Identification of these biomarkers would assist not only in distinguishing NSCLC into its major subtypes - Adenocarcinoma and Squamous Cell Carcinoma, but also in developing targeted therapy. Medical practitioners use one or more types of omic data to identify these biomarkers, copy number variation (CNV) being one such type. CNV provides a measure of genomic instability, which is considered a hallmark of carcinoma. However, the CNV data has not received much attention for biomarker identification. This paper aims to identify biomarkers for NSCLC using CNV data. METHODS An eXplainable AI (XAI)-driven L1-regularized deep learning architecture, XL1R-Net, is proposed that introduces a novel modification of the standard L1-regularized gradient descent algorithm to arrive at an improved deep neural classifier for NSCLC subtyping. Further, XAI-based feature identification has been used to leverage the trained classifier to uncover a set of twenty NCSLC-relevant biomarkers. RESULTS The identified biomarkers are evaluated based on their classification performance and clinical relevance. Using Multilayer Perceptron (MLP)-based model, a classification accuracy of 84.95% using 10-fold cross-validation is achieved. Moreover, the statistical significance test on the classification performance also revealed the superiority of the MLP model over the competitive machine learning models. Further, the publicly available Drug-Gene Interaction Database reveals twelve of the identified biomarkers as potentially druggable. The K-M Plotter tool was used to verify eighteen of the identified biomarkers with a high probability of predicting NSCLC patients' likelihood of survival. While nine of the identified biomarkers confirm the recent literature, five find mention in the OncoKB Gene List. CONCLUSION A set of seven novel biomarkers that have not been reported in the literature could be investigated for their potential contribution towards NSCLC therapy. Given NSCLC's genetic diversity, using only one omics data type may not adequately capture the tumor's complexity. Multiomics data and its integration with other sources will be examined in the future to better understand NSCLC heterogeneity.
Collapse
Affiliation(s)
- Kountay Dwivedi
- Department of Computer Science, University of Delhi, Delhi, India.
| | - Ankit Rajpal
- Department of Computer Science, University of Delhi, Delhi, India.
| | - Sheetal Rajpal
- Department of Computer Science, Dyal Singh College, Delhi, India.
| | - Virendra Kumar
- Department of Nuclear Magnetic Resonance, All India Institute of Medical Sciences, New Delhi, India.
| | - Manoj Agarwal
- Department of Computer Science, Hans Raj College, University of Delhi, Delhi, India.
| | - Naveen Kumar
- Department of Computer Science, University of Delhi, Delhi, India.
| |
Collapse
|
45
|
Vescio M, Bulloni M, Pelosi G, Pattini L. Lack of imbalance between the master regulators TTF1/NKX2-1 and ΔNp63/p40 implies adverse prognosis in non-small cell lung cancer. Sci Rep 2024; 14:2467. [PMID: 38291083 PMCID: PMC10827720 DOI: 10.1038/s41598-024-52776-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
The transcription factors TTF1/NKX2-1 and ΔNp63/p40 are the counterposed molecular markers associated with the main Non-Small Cell Lung Cancer subtypes: TTF1 for adenocarcinoma, p40 for squamous cell carcinoma. Although they generally display a mutually exclusive expression, some exceptions exist simultaneously lacking or (very rarely) expressing both markers, either pattern being associated to poor prognosis. Hence, we quantitatively analyzed the relationship between their coordinated activity and prognosis. By analyzing the respective downstream transcriptional programs of the two genes, we defined a simple quantitative index summarizing the amount of mutual exclusivity between their activities, called Mean Absolute Activity (MAA). Systematic analysis of the MAA index in a dataset of 1018 NSCLC samples replicated on a validation dataset of 275 showed that the loss of imbalance between TTF-1 and p40 corresponds to a steady, progressive reduction in both overall and recurrence-free survival. Coherently, samples correspondent to more balanced activities were enriched for pathways related to increased malignancy and invasiveness. Importantly, multivariate analysis showed that the prognostic significance of the proposed index MAA is independent of other clinical variables including stage, sex, age and smoke exposure. These results hold irrespectively of tumor morphology across NSCLC subtypes, providing a unifying description of different expression patterns.
Collapse
Affiliation(s)
- Martina Vescio
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
- CardioTech, IRCCS Centro Cardiologico Monzino, Milan, Italy
| | - Matteo Bulloni
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Inter-Hospital Pathology Division, IRCCS MultiMedica, Milan, Italy
| | - Linda Pattini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy.
- CardioTech, IRCCS Centro Cardiologico Monzino, Milan, Italy.
| |
Collapse
|
46
|
Moes-Sosnowska J, Szpechcinski A, Chorostowska-Wynimko J. Clinical significance of TP53 alterations in advanced NSCLC patients treated with EGFR, ALK and ROS1 tyrosine kinase inhibitors: An update. Tumour Biol 2024; 46:S309-S325. [PMID: 37840519 DOI: 10.3233/tub-230034] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
The development of targeted therapies for non-small cell lung cancer (NSCLC), such as the epidermal growth factor receptor (EGFR), anaplastic lymphoma receptor tyrosine kinase (ALK), and ROS proto-oncogene 1 (ROS1), has improved patients' prognosis and significantly extended progression-free survival. However, it remains unclear why some patients do not benefit from the treatment as much or have a rapid disease progression. It is considered that, apart from the oncogenic driver gene, molecular alterations in a number of caretaker and gatekeeper genes significantly impact the efficacy of targeted therapies. The tumor protein 53 (TP53) gene is one of the most frequently mutated genes in NSCLC. To date, numerous studies have investigated the influence of various TP53 alterations on patient prognosis and responsiveness to therapies targeting EGFR, ALK, or ROS1. This review focuses on the latest data concerning the role of TP53 alterations as prognostic and/or predictive biomarkers for EGFR, ALK, and ROS1 tyrosine kinase inhibitors (TKIs) in advanced NSCLC patients. Since the presence of TP53 mutations in NSCLC has been linked to its decreased responsiveness to EGFR, ALK, and ROS1 targeted therapy in most of the referenced studies, the review also discusses the impact of TP53 mutations on treatment resistance. It seems plausible that assessing the TP53 mutation status could aid in patient stratification for optimal clinical decision-making. However, drawing meaningful conclusions about the clinical value of the TP53 co-mutations in EGFR-, ALK- or ROS1-positive NSCLC is hampered mainly by an insufficient knowledge regarding the functional consequences of the TP53 alterations. The integration of next-generation sequencing into the routine molecular diagnostics of cancer patients will facilitate the detection and identification of targetable genetic alterations along with co-occurring TP53 variants. This advancement holds the potential to accelerate understanding of the biological and clinical role of p53 in targeted therapies for NSCLC.
Collapse
Affiliation(s)
- Joanna Moes-Sosnowska
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Adam Szpechcinski
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| |
Collapse
|
47
|
Zeng P, Wang F, Zhang J, Ur Rashid H, Li X, Zhang P, Luo Y, Wu X. Integrating network pharmacology and experimental verification to investigate the pharmacological mechanisms of Buzhong Yiqi decoction in the treatment of non-small cell lung cancer. Chem Biol Drug Des 2024; 103:e14414. [PMID: 38230796 DOI: 10.1111/cbdd.14414] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024]
Abstract
Among all types of cancers, non-small cell lung cancer (NSCLC) exhibits the highest mortality rate with a five-year survival rate below 17% for patients. The Buzhong Yiqi decoction (BZYQD), traditional Chinese medicine (TCM) formula, has been reported to exhibit clinical efficacy in the treatment of NSCLC. Nevertheless, the underlying molecular mechanism remains elusive. This study aimed to assess the mechanistic actions exerted by BZYQD against NSCLC using network pharmacological analysis and experimental validation. The public databases were searched for active compounds in BZYQD, their potential targets, and NSCLC-related targets. The protein-protein interaction (PPI) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to predict the core targets and signaling pathways of BZYQD against NSCLC. After screening, this study validated the results of predictions through in vitro experiments and public databases. We found 192 common targets between BZYQD and NSCLC. KEGG analysis showed that the anti-NSCLC effects of BZYQD were mediated through the PI3K-AKT signaling pathway. The results of in vitro experiment indicated that BZYQD could inhibit cell viability and proliferation of A549 and H1299 cells apart from inducing cell apoptosis. In addition, western blot results substantiated that BZYQD could treat NSCLC by inhibiting the activation of the PI3K-AKT signaling pathway. The current study investigated the pharmacological mechanism of BZYQD against NSCLC via network pharmacology and in vitro analyses. Overall, the results revealed that BZYQD could be a promising therapeutic agent for the treatment of NSCLC in the future. Still, more experimental investigations are needed to confirm the applicability of BZYQD for clinical trials.
Collapse
Affiliation(s)
- Panke Zeng
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Feng Wang
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jianing Zhang
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haroon Ur Rashid
- Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Pelotas, Brazil
| | - Xin Li
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunru Luo
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyu Wu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
48
|
Mendoza RP, Chen‐Yost HI, Wanjari P, Wang P, Symes E, Johnson DN, Reeves W, Mueller J, Antic T, Biernacka A. Lung adenocarcinomas with isolated TP53 mutation: A comprehensive clinical, cytopathologic and molecular characterization. Cancer Med 2024; 13:e6873. [PMID: 38164123 PMCID: PMC10824142 DOI: 10.1002/cam4.6873] [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: 10/11/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND TP53 mutation is present in about 50.8% of lung adenocarcinomas, frequently in combination with other genetic alterations. However, a rare subset harbors the TP53 mutation alone. METHODS Next-generation sequencing was performed in 844 lung adenocarcinomas diagnosed by fine needle aspiration. Fourteen cases (1.7%) showed isolated TP53 alteration and were subjected to a comprehensive analysis. RESULTS The average age at diagnosis was 65.7 years (range 48-79); 9 males and 5 females. All were smokers with an average pack-year of 40.7 (range 10-70). Ten had metastases, mostly in the brain (n = 4) and pleura (n = 4). After a follow-up period of up to 102 months, 9 died, 3 were alive free of disease, 1 was alive with disease, and 1 was lost to follow-up. The median survival was 12.2 months. Most tumors exhibited poor differentiation, composed of solid sheets with moderate to severe atypia, increased mitotic activity, and necrotic background. Half were positive for TTF-1 and showed p53 overexpression. PD-L1 was positive in 5 cases. Most alterations were missense mutations in exons 5-8, and this mutation type was associated with p53 overexpression. Tumors with combined missense mutation and truncated protein had higher PD-L1 expression along with a trend towards an increase in tumor mutational burden (TMB). CEBPA deletion of undetermined significance was the most common copy number alteration. CONCLUSION Isolated TP53 mutation was seen in association with smoking, high-grade cytomorphologic features, adverse prognosis, and recurrent CEBPA deletions. These tumors tend to have strong PD-L1 expression and high TMB, suggesting potential benefit from immune checkpoint inhibitors. Hence, the recognition of this molecular group has prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Rachelle P. Mendoza
- Department of PathologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | | | - Pankhuri Wanjari
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| | - Peng Wang
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| | - Emily Symes
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| | - Daniel N. Johnson
- Department of PathologyOSF Little Company of Mary Medical CenterEvergreen ParkIllinoisUSA
| | - Ward Reeves
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| | - Jeffrey Mueller
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| | - Tatjana Antic
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| | - Anna Biernacka
- Department of PathologyThe University of Chicago HospitalsChicagoIllinoisUSA
| |
Collapse
|
49
|
Jiang T, Lu Y, Yang W, Xu J, Zhu M, Huang Y, Bao F, Zheng S, Li Y. To Explore the Mechanism of Maiwei Dihuang Decoction in the Treatment of Non-small Cell Lung Cancer based on Network Pharmacology Combined with LC-MS. Curr Comput Aided Drug Des 2024; 20:590-597. [PMID: 37612858 DOI: 10.2174/1573409920666230823161355] [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: 11/22/2022] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023]
Abstract
OBJECTIVE To explore the mechanism of Maiwei Dihuang decoction in the treatment of non-small cell lung cancer (NSCLC) by using network pharmacology and LC-MS technology. METHODS The effective components in Maiwei Dihuang decoction were detected by liquid chromatography- mass spectrometry (LC-MS). Use the SuperPred database to collect the relevant targets of the active ingredients of Mai Wei Di Tang, and then collect the relevant targets of nonsmall cell lung cancer from GeneCards, DisgenNET and OMIM databases. On this basis, PPI network construction, GO enrichment analysis and KEGG pathway annotation analysis were carried out for target sites. Finally, AutoDock Vina is used for molecular docking. RESULTS We further screened 16 effective Chinese herbal compounds through LC-MS combined with ADME level. On this basis, we obtained 77 core targets through protein interaction network analysis. Through GO, KEGG analysis and molecular docking results, we finally screened out the potential targets of Maiwei Dihuang Decoction for NSCLC: TP53, STAT3, MAPK3. CONCLUSION Maiwei Dihuang decoction may play a role in the treatment of NSCLC by coregulating TP53/STAT3/MAPK3 signal pathway.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Yang Lu
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Wanzhi Yang
- Department of Pharmacy, Anqing First People's Hospital of Anhui Medical University, Anhui Province, China
| | - Jinhong Xu
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Mingxing Zhu
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Yong Huang
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Fang Bao
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Shengqi Zheng
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| | - Yongxia Li
- Department of Pharmacy, Navy Anqing Hospital, Anqing, Anhui Province, China
| |
Collapse
|
50
|
Choi DH, Jung HA, Park S, Sun JM, Ahn JS, Ahn MJ, Lee SH. Effectiveness and safety of amivantamab in EGFR exon 20 insertion (E20I) mutations in non-small cell lung cancer (NSCLC). Transl Lung Cancer Res 2023; 12:2448-2459. [PMID: 38205202 PMCID: PMC10775014 DOI: 10.21037/tlcr-23-643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
Background In non-small cell lung cancer (NSCLC), the epidermal growth factor receptor (EGFR) mutation is a representative oncogenic driver mutation. Only about 12% of EGFR mutation patients have the exon 20 insertion mutation, which is the third most frequent mutation among EGFR mutation NSCLC. Amivantamab, an EGFR and MET proto-oncogene, receptor tyrosine kinase (MET) bispecific antibody, was approved for NSCLC patients with the EGFR exon 20 insertion (E20I) mutation. In this study, we described the real-world, single-center efficacy and safety data of amivantamab in E20I mutation patients. Methods This study included metastatic NSCLC patients with EGFR E20I mutations. From January 2018 to June 2022, patients with EGFR E20I mutations who were treated with amivantamab were analyzed at Samsung Medical Center as part of the clinical trial or the early access program (EAP). We collected the patients' characteristics [age, sex, smoking history, location of mutation, sites of metastasis, programmed death-ligand 1 (PD-L1) expression status, etc.] and analyzed progression-free survival (PFS) and overall survival (OS) stratified by PD-L1 expression status, co-mutation such as tumor protein p53 (TP53), and metastasis sites. Results A total of 42 patients were analyzed, of which 16 patients were enrolled in the phase 1 study, and 26 patients received amivantamab through EAP. There were 14 (33%) patients with partial remission, 18 (43%) patients with stable disease, and 10 (24%) patients with disease progression. The objective response rate (ORR) was 33%, and the disease control rate (DCR) was 76%. PFS was analyzed by dividing the near and far loop for 31 patients whose mutation location was known. The two groups had no statistically significant difference in PFS [median (range): 11.8 (2.3-21.3) vs. 11.3 (3.4-19.2) months, P=0.69]. For 29 patients with TP53 mutation data, there was no significant difference in PFS between the two groups [median (range): 5.9 (0-18.0) vs. 12.6 (6.9-18.3) months, P=0.11]. When analyzing PFS in 37 patients with PD-L1 expression data, PD-L1 (+) patients showed a poor prognosis [median (range): 11.3 (5.0-17.6) vs. 19.5 (5.3-33.7) months, P=0.04; hazard ratio (HR), 0.44; 95% confidence interval (CI): 0.20-0.98]. Conclusions The efficacy of amivantamab was confirmed for the real-world population for EGFR E20I-mutated NSCLC. PD-L1 status could be a poor predictive factor, which should be further validated.
Collapse
Affiliation(s)
- Dae-Ho Choi
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|