1
|
Zhang T, Yang J, Li S, Shi X, Yang J. Ovarian squamous cell carcinoma: clinicopathological features, prognosis and immunotherapy outcomes. J Gynecol Oncol 2025; 36:36.e54. [PMID: 39791511 DOI: 10.3802/jgo.2025.36.e54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/21/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVE To explore the characteristics and survival outcomes of ovarian squamous cell carcinoma (SCC) and the treatment effectiveness of immune checkpoint inhibitors (ICIs). METHODS Patients diagnosed with ovarian SCC at Peking Union Medical College Hospital between January 2000 and September 2023 were included. Overall survival (OS) and progression-free survival (PFS) were analyzed using the Kaplan-Meier method. Univariate and multivariate analysis of OS were performed using the Cox proportional hazards model. RESULTS A total of 42 patients were included, with a median age of 51.5 years (range, 23-74). The majority had SCC arising from teratomas (54.8%), followed by endometriosis (14.3%) and Brenner's tumor (2.4%). Patients undergoing molecular testing exhibited a median tumor mutation burden (TMB) of 10.00 mutations/Mb (range, 7.28-46.86), predominantly featuring PIK3CA mutations. Thirty-eight patients (90.5%) received adjuvant chemotherapy. The median OS was 42.0 months, with the 1- and 5-year OS rates were 73.7% and 48.7%, respectively. And the median PFS was 26.9 months, with the 1- and 5-year PFS rates were 57.5% and 43.8%, respectively. Five patients underwent first-line postoperative adjuvant therapy combining ICIs with chemotherapy. During the 9.5 to 25.1 months follow-up, 4 patients showed no evidence of disease, while 1 relapsed and received treatment. Late-stage disease and younger age at diagnosis were associated with worse survival outcomes. CONCLUSION The prognosis for ovarian SCC remains unfavorable. The stage and age were prognostic predictors for survival. ICIs may be beneficial for patients with ovarian SCC, particularly those with a high TMB.
Collapse
Affiliation(s)
- Tianyu Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Jie Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China.
| | - Sijian Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Xiaohua Shi
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| |
Collapse
|
2
|
Chen XS, Chen F, He SJ, Chen YY, Chi BT, Huang WY, Wei Y, Zhao CY, Song C, He RQ, Chen G, Kong JL, Lu HP. Elevated expression of ANAPC1 in lung squamous cell carcinoma: clinical implications and mechanisms. Future Sci OA 2025; 11:2482487. [PMID: 40139913 PMCID: PMC11951694 DOI: 10.1080/20565623.2025.2482487] [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: 11/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
AIM To investigate the comprehensive expression levels and possible molecular mechanisms of Anaphase Promoting Complex Subunit 1 (ANAPC1) in lung squamous cell carcinoma (LUSC). METHODS Data from 2,031 samples were combined to evaluate ANAPC1 mRNA levels, and 118 samples were collected for immunohistochemical (IHC) analysis. High-expression co-expressed genes (HECEGs) associated with ANAPC1 were analyzed for signaling pathways. Clinical significance, immune computations, and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) validation of ANAPC1's role in LUSC were assessed. Molecular docking evaluated binding affinity with potential therapeutics. RESULTS ANAPC1 mRNA was significantly upregulated in LUSC (SMD = 1.97, 95% CI [1.26-2.67]). Protein-level analysis confirmed this upregulation (p < 0.001). Most HECEGs associated with ANAPC1 were enriched in cell cycle pathways. Higher ANAPC1 expression correlated with poorer survival in LUSC patients (HR = 1.11, 95% CI: 1-1.49). ANAPC1 expression was higher in males and N1-stage vs. females and N0-stage; lower in grade I vs. II/III. Overexpression reduces immune cell infiltration and immunotherapy effectiveness, while knockdown inhibits cell proliferation. Drug sensitivity and docking analyses identified tenovin-1, carboxyatractyloside, and phycocyanobilin as potential antitumor agents targeting ANAPC1. CONCLUSION The elevated expression of ANAPC1 might play a role in LUSC advancement and progression through its participation in cell growth-related pathways.
Collapse
Affiliation(s)
- Xiao-Song Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Feng Chen
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shu-Jia He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yi-Yang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bang-Teng Chi
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wan-Ying Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yue Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chun-Yan Zhao
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chang Song
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rong-Quan He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jin-Liang Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hui-Ping Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
3
|
García-Pardo M, García de Herreros M, Laguna JC, Gorría T, Lage Y, Gómez A, Olmedo ME, Garrido P, Albarran-Artahona VM, Arcocha A, Teixido C, Reguart N, Navarro A, Auclin E, Nahorski MS, Howarth K, Planchard D, Besse B, Leighl NB, Mezquita L. Brief Report: Real-World Clinical Utility of Next-Generation Sequencing of Circulating Tumor DNA for Patients With Advanced Lung Squamous Cell Carcinoma (SQUIN). Clin Lung Cancer 2025; 26:e300-e305.e2. [PMID: 40064572 DOI: 10.1016/j.cllc.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 05/24/2025]
Affiliation(s)
- Miguel García-Pardo
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Marta García de Herreros
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan Carlos Laguna
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Teresa Gorría
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Yolanda Lage
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Ana Gómez
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Mª Eugenia Olmedo
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Pilar Garrido
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Víctor Manuel Albarran-Artahona
- Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Thoracic Group and International Center for Thoracic Cancers (CICT), Gustave Roussy, Villejuif, France
| | - Ainara Arcocha
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Cristina Teixido
- Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Pathology Department, Hospital Clinic y Provincial de Barcelona, Barcelona, Spain
| | - Noemí Reguart
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alejandro Navarro
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Edouard Auclin
- Oncology Department, HEGP - Hopital European Georges-Pompidou - AP-HP, Paris, France
| | | | | | - David Planchard
- Department of Medical Oncology, Thoracic Group and International Center for Thoracic Cancers (CICT), Gustave Roussy, Villejuif, France; Faculty of Medicine, Paris-Saclay University, Paris, France
| | - Benjamin Besse
- Department of Medical Oncology, Thoracic Group and International Center for Thoracic Cancers (CICT), Gustave Roussy, Villejuif, France; Faculty of Medicine, Paris-Saclay University, Paris, France
| | - Natasha B Leighl
- Medical Oncology Department, Princess Margaret Cancer Center, Toronto, Canada
| | - Laura Mezquita
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted therapies in Solid Tumors, Institut de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Huang C, Yang H, Wang H, Sheng S, Chen L. HDCA6 suppresses GATA6 expression to enhance cellular growth and migration in lung squamous cell carcinoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167757. [PMID: 40044063 DOI: 10.1016/j.bbadis.2025.167757] [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: 10/31/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Lung squamous cell carcinoma (LUSC) exhibits a significant mortality rate and lacks effective targeted therapies. The GATA-binding factor 6 (GATA6), a pivotal regulator of lung development, undergoes reduced expression in LUSC and correlates with its metastasis and prognosis. However, the regulatory mechanisms underlying the down-regulation of GATA6 in LUSC remain elusive. HDAC6 emerges as a promising therapeutic target in preclinical cancer models. Nevertheless, its role in LUSC progression remains unexplored. Furthermore, the regulatory impact of HDAC6 on GATA6 expression needs clarification. The purpose of this work is to investigate HDAC6's involvement and regulatory mechanisms on the expression of GATA6 in LUSC. METHODS The impacts of HDCA6 on the expression of GATA6 were assessed using qPCR, Western blot and CHIP assays. The tumorigenic capacity of HDAC6 in promoting the growth and migration of LUSC cell were determined through CCK8, EdU incorporation, Transwell, and xenograft tumor models. Immunohistochemistry assays were employed to detected expression of HDAC6 in tissue microarray of LUSC. RESULTS A pan-HDACs inhibitor Trichostatin A and an HDAC6-specific inhibitor CAY10603 up-regulate the expression of GATA6, whereas HDAC6 overexpression down-regulates GATA6 level. Overexpression of HDAC6 promotes cell proliferation and migration in LUSC, while inhibition of HDAC6 significantly suppresses LUSC cell growth. And, GATA6 overexpression reverses HDAC6-mediated elevated growth and migration of LUSC cells. Compared to normal tissues, LUSC tissues exhibit elevated levels of HDAC6 expression, which were correlated with poor prognosis of LUSC patients. CONCLUSION Targeting HDAC6/GATA6 pathway may offer promising prospects for developing of novel therapeutic strategies against LUSC.
Collapse
Affiliation(s)
- Changhua Huang
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hua Yang
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hailong Wang
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Shouyin Sheng
- Department of Surgery 2, Jiujiang Third People's Hospital, Jiujiang, Jiangxi 332099, China.
| | - Limin Chen
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
5
|
Song S, Lu R, Chen Y, Feng Y. PCDHGA10 as a Potential Biomarker of Lung Squamous Cell Carcinoma Based on Bioinformatics and Experimental Verification. Mol Biotechnol 2025; 67:2002-2022. [PMID: 38727881 DOI: 10.1007/s12033-024-01178-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/15/2024] [Indexed: 04/10/2025]
Abstract
Procalcitonin gamma subfamily A, 10 (PCDHGA10) is a member of the procalcitonin gamma gene cluster, which is associated with neuronal synapse development. However, there are lack of studies on the role and potential prognostic value of PCDHGA10 in lung squamous cell carcinoma (LUSC). We analyzed the RNAseq data of PCDHGA10 to compare their expression differences. Then survival analysis, tumor microenvironment (TME) analysis, and mutation analysis were carried out. Additionally, we performed gene ontology (GO) and kyoto gene encyclopedia (KEGG) enrichment analyses to explore potential signal pathways. PCDHGA10 protein expression was evaluated using immunohistochemistry (IHC) on tissue microarrays (HLugS180Su02). By microarray analysis and database analysis, we found that PCDHGA10 was significantly highly expressed in LUSC. Sufferers with elevated PCDHGA10 levels exhibited a worse prognosis, according to the survival analysis. The PCDHGA10 mutation was also linked to LUSC patient prognosis. Besides, PCDHGA10 was closely related to tumor immune cell infiltration and immune checkpoints. In conclusion, PCDHGA10 is expected to become a new molecular marker for LUSC.
Collapse
Affiliation(s)
- Shuming Song
- Xinjiang Key Laboratory of Oncology, Cancer Hospial Affiliated to Xinjiang Medical University, Urumqi, China
- People's Hospital of Bachu County, Xinjiang, Kashgar, China
| | - Ruijiao Lu
- Clinical Laboratory Center, Cancer Hospial Affiliated to Xinjiang Medical University, Urumqi, China
| | - Yuanya Chen
- General Surgery Department, People's Hospital of Bachu County, Xinjiang, Kashgar, China
| | - Yangchun Feng
- Clinical Laboratory Center, Cancer Hospial Affiliated to Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
6
|
He A, Huang Z, Chen X, Qi K, Zhang S, Li F, Lu H, Wang J, Peng J, Song C. Decoding the role of lipid metabolism in NSCLC: From macrophage subtype identification to prognostic model development. FASEB J 2025; 39:e70467. [PMID: 40277347 DOI: 10.1096/fj.202500124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/07/2025] [Indexed: 04/26/2025]
Abstract
Lipid metabolism plays a pivotal role in shaping the tumor microenvironment, particularly by influencing macrophage function. This study aimed to identify lipid-associated macrophage (LAM) marker genes involved in the onset and progression of non-small cell lung cancer (NSCLC) through integrated single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) analyses. Mutation and RNA-seq data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were analyzed to explore the relationship between lipid metabolism pathways and NSCLC progression. scRNA-seq analysis revealed macrophage subtypes closely associated with lipid metabolism, with three key marker genes-S100A10, HLA-DMB, and CTSL-identified as predictive factors for patient prognosis. A prognostic risk scoring model was constructed and validated using survival analysis and ROC curves, demonstrating high accuracy in stratifying NSCLC patients by risk. Further in vivo experiments using subcutaneous tumor xenografts and lung metastasis models showed that S100A10 and CTSL promoted tumor growth and metastasis, while HLA-DMB inhibited these processes. Immune infiltration analysis highlighted the immunological relevance of the identified marker genes, providing insights into their functional roles. This study underscores the critical influence of LAMs in NSCLC progression and highlights a robust prognostic model that offers potential therapeutic targets for improving patient outcomes.
Collapse
Affiliation(s)
- Aoxiao He
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xianglai Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kai Qi
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shan Zhang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Li
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jinhua Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chao Song
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
García-Hernández L, Dai L, Rodríguez-Ulloa A, Yi Y, González LJ, Besada V, Li W, Perea SE, Perera Y. Time- and dose-dependent effects of CIGB-300 on the proteome of lung squamous cell carcinoma. Biol Chem 2025:hsz-2024-0149. [PMID: 40261874 DOI: 10.1515/hsz-2024-0149] [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: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Proteome-wide scale in a dose - and time-depending setting is crucial to fully understand the pharmacological mechanism of anticancer drugs as well as identification of candidates for drug response biomarkers. Here, we investigated the effect of the CIGB-300 anticancer peptide at IC50 and IC80 doses during 1 and 4 h of treatment on the squamous lung cancer cell (NCI-H226) proteome. An overwhelming dose-dependent inhibitory effect with minor up-regulated proteins was observed by increasing CIGB-300 dose level. Functional enrichment was also CIGB-300 dose-dependent with common or exclusively regulated proteins in each dose and time settings. A protein core involving small molecule biosynthesis, aldehyde metabolism and metabolism of nucleobases was regulated irrespectively to the dose or the treatment time. Importantly, a group of proteins linked to NSCLC tumor biology, poor clinical outcome and some Protein Kinase CK2 substrates, were significantly regulated by treating with both CIGB-300 doses. Likewise, we observed a consistent downregulation of different proteins that had been already reported to be inhibited by CIGB-300 in lung adenocarcinoma and acute myeloid leukemia. Overall, our proteomics-guided strategy based on time and drug dose served to uncover novel clues supporting the CIGB-300 cytotoxic effect and also to identify putative pharmacodynamic biomarkers in NSCLC.
Collapse
Affiliation(s)
- Liudy García-Hernández
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Lingfeng Dai
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| | - Arielis Rodríguez-Ulloa
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Ying Yi
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| | - Luis J González
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Vladimir Besada
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Wen Li
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| | - Silvio E Perea
- Department of Pharmaceuticals, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Playa, Havana 10600, Cuba
| | - Yasser Perera
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| |
Collapse
|
8
|
Zha Z, Liu C, Yan M, Chen C, Yu C, Chen Y, Zhou C, Li L, Li YC, Yamaguchi H, Ye L, Liu T, Wang YN, Lee HH, Yang WH, Chan LC, Ke B, Hsu JL, Ding L, Ji D, Pan P, Meng Y, Pu Y, Liu L, Hung MC. RNase1-driven ALK-activation is an oncogenic driver and therapeutic target in non-small cell lung cancer. Signal Transduct Target Ther 2025; 10:124. [PMID: 40246819 PMCID: PMC12006399 DOI: 10.1038/s41392-025-02206-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/18/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Targeted therapy has achieved significant success in the treatment of non-small cell lung cancer (NSCLC), particularly in patients harboring common oncogenic driver mutations such as EGFR, KRAS, and ALK rearrangement. However, ~35-50% of NSCLC patients without tyrosine kinase mutation or rearrangement (non-mutated) cannot benefit from these targeted treatments, highlighting the urgent need for novel therapeutic strategies for this patient population. In this study, we report a non-canonical role of human secretory ribonuclease 1 (RNase1), which binds to and activates wild-type ALK in lung cancer cells, thereby triggering its downstream signaling pathway. RNase1-driven ALK-activation (RDAA) cells exhibit enhanced cell proliferation, migration, and colony formation. Additionally, RDAA facilitates tumor formation in fibroblast models, further underscoring its oncogenic potential in vivo. Importantly, RDAA lung cancer cells exhibit marked sensitivity to FDA-approved ALK inhibitors. Tumor growth suppression and survival were substantially improved in both RDAA-positive NSCLC cell line-derived and patient-derived xenograft tumor models treated with ALK inhibitors. Monoclonal antibodies against RNase1 and phosphorylated-ALK were used to analyze two different human NSCLC tissue cohorts by immunohistochemical staining identified 10.4% (5/48) and 8.5% (100/1173) patients who were RDAA positive, respectively. Notably, among the nine RDAA-positive NSCLC patients who accepted ALK inhibitor treatment, five achieved objective response including two who experienced complete response (CR). Together, the current study identifies RDAA as an oncogenic driver and proposes an effective targeted therapy strategy for non-mutated NSCLC patients.
Collapse
Affiliation(s)
- Zhengyu Zha
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Chunxiao Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meisi Yan
- Department of Pathology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Cong Chen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Cheng Yu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Yaohui Chen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Chenhao Zhou
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Liver Cancer and Transplant, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Lu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi-Chuan Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiro Yamaguchi
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Leiguang Ye
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tong Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen-Hao Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Baozhen Ke
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Dong Ji
- Betta Pharmaceuticals Co. Ltd, Hangzhou, China
| | - Peng Pan
- Betta Pharmaceuticals Co. Ltd, Hangzhou, China
| | - Yiran Meng
- Hangzhou Repugene Technology Co., Ltd, Hangzhou, China
| | - Yue Pu
- Hangzhou Repugene Technology Co., Ltd, Hangzhou, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China.
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Wang D, Wang Y, Peng Y, Peng L. Utilizing multi-omics analysis, a new signature has been identified and validated for predicting prognosis and response to immunotherapy in lung squamous cell carcinoma, which is based on tumor mutation burden. Discov Oncol 2025; 16:539. [PMID: 40240626 PMCID: PMC12003222 DOI: 10.1007/s12672-025-02166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Immunotherapy is used extensively in treating non-small cell lung cancer (NSCLC) patients. Nevertheless, in contrast to lung adenocarcinoma (LUAD), the endeavors to develop effective targeted treatments for lung squamous cell carcinoma (LUSC) have not yielded positive outcomes. Hence, it is crucial to discover biomarkers for immunotherapy and investigate more potent treatments, which is an immediate requirement for individuals with LUSC. The LUSC somatic mutation data were obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Multivariate analysis was performed to create a signature related to tumor mutation burden (TMB). Next, we utilized the CIBERSORT algorithm to assess the correlation between TMB and immune infiltrates. Additionally, we identified prognostic immune cells of LUSC through Kaplan-Meier analysis. The TCGA and ICGC cohorts covered a combined total of 11 genes that were frequently mutated. SYNE1 and TTN mutation correlated with an increased TMB and suggested a positive clinical outlook. A TMB-related signature (SYNE1 and TTN) was constructed based on this. The outlook for the high-risk group in LUSC was considerably poorer than the low-risk group (p = 0.004). In LUSC, there was a correlation between the TMB-related signature and immune infiltrates, and a positive response to anti-PD-L1 therapy was observed in individuals with low-risk scores. Furthermore, based on Kaplan-Meier analysis, plasma cells were identified as predictive immune cells in LUSC samples. In conclusion, the GSEA examination demonstrated that the TMB-associated signature stimulated immune system-related signaling pathways. To sum up, the TMB-associated signature could be marker to anticipate the immune reaction in individuals with LUSC.
Collapse
Affiliation(s)
- Dongguang Wang
- Department of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, China
- The Innovation and Entrepreneurship Education Center for Agricultural Biotechnology of Hunan Province, Loudi, 417000, China
| | - Yan Wang
- Department of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, China
- The Innovation and Entrepreneurship Education Center for Agricultural Biotechnology of Hunan Province, Loudi, 417000, China
| | - Yiqun Peng
- Department of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, China
- The Innovation and Entrepreneurship Education Center for Agricultural Biotechnology of Hunan Province, Loudi, 417000, China
| | - Liang Peng
- Department of Nephrology, The Second Affiliated Hospital of University of South China, Hengyang, 421001, China.
| |
Collapse
|
10
|
Zhong S, Börgeling Y, Zardo P, Jonigk D, Borlak J. Comprehensive transcriptome, miRNA and kinome profiling identifies new treatment options for personalized lung cancer therapy. Clin Transl Med 2025; 15:e70177. [PMID: 39995112 PMCID: PMC11850761 DOI: 10.1002/ctm2.70177] [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: 09/13/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Basic research identified oncogenic driver mutations in lung cancer (LC). However, <10% of patients carry driver mutations. Thus, most patients are not recommended for first-line kinase inhibitor (KI)-based therapies. Through enabling technologies and bioinformatics, we gained deep insight into patient-specific signalling networks which permitted novel KI-based treatment options in LC. METHODS We performed molecular pathology, transcriptomics and miRNA profiling across 95 well-characterized LC patients. We confirmed results based on cross-linked immunoprecipitation-sequencing data, and used N = 524 adeno- and 497 squamous cell carcinomas as validation sets. We employed the PamGene platform to identify aberrant kinases, validated the results by evaluating independent siRNA and CRISPR-mediated mRNA knockdown studies in human LC cell lines. RESULTS Transcriptomics revealed 439, 1240, 383 and 320 significantly upregulated genes, respectively, for adeno-, squamous, neuroendocrine and metastatic cases, and there are 1092, 1477, 609 and 1267 downregulated DEGs. Based on gene enrichment analysis and experimentally validated miRNA-gene interactions, we constructed regulatory networks specific for adeno-, squamous, neuroendocrine and metastatic LC. Molecular profiling discovered 137 significantly upregulated kinases (range 2-26-fold) of which 65 and 72, respectively, are tyrosine and serine-threonine kinases while 6 kinases carry driver mutations. Meanwhile, there are 21 kinases commonly upregulated irrespective of the histological type of LC. Bioinformatics decoded networks in which kinases function as master regulators. Typically, the networks consisted of 14, 9, 16 and 19 highly regulated kinases in adeno-, squamous, neuroendocrine and metastatic LC. Inhibition of kinases which function as master regulators disrupted the signalling networks, and their gene knock-down studies confirmed inhibition of cell proliferation in a panel of human LC cell lines. Additionally, the proposed molecular profiling enables KI-based therapies in patients with acquired drug resistance. CONCLUSIONS Our study broadens the perspective of KI-based therapies in LC, and we propose a framework to overcome acquired drug resistance.
Collapse
Affiliation(s)
- Shen Zhong
- Centre for Pharmacology and ToxicologyHannover Medical SchoolHannoverGermany
| | | | - Patrick Zardo
- Clinic for Cardiothoracic and Transplantation SurgeryHannover Medical SchoolHannoverGermany
| | - Danny Jonigk
- Institute for PathologyHannover Medical SchoolHannoverGermany
| | - Jürgen Borlak
- Centre for Pharmacology and ToxicologyHannover Medical SchoolHannoverGermany
| |
Collapse
|
11
|
Wang H, Liu C, Jiang C, Zhang Y, Zhao X, Jia Z, Huo J, Yang J. GRHL3 drives radiotherapy resistance and blocks the anti-tumor response of NK and CD4 + T cells in lung squamous cell carcinoma via RNF2. Biochem Pharmacol 2025; 233:116784. [PMID: 39880318 DOI: 10.1016/j.bcp.2025.116784] [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: 06/18/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Grainyhead-like protein 3 homolog (GRHL3) has been identified as a top transcription factor associated with keratinization in lung squamous cell carcinoma (LUSC). We designed this study to elucidate the function of GRHL3 in radioresistance in LUSC and the mechanism involved. Transcriptome differences between radioresistant and parental cells were analyzed to identify the hub transcription factor. GRHL3 expression was overexpressed in radioresistant cells relative to parental cells, and the knockdown of GRHL3 conferred sensitivity to radioresistant LUSC cells, induced DNA damage, inhibited cell survival, and reduced tumor load in mice. GRHL3 promoted ring finger protein 2 (RNF2) transcription by binding to the RNF2 promoter. GRHL3 induced a radioresistant phenotype in parental cells and led to compromised anti-tumor immune responses of CD4+ T cells and NK cells. The GRHL3-promoted tumor progression was reversed by the knockdown of RNF2. The DNA methylation of GRHL3 was reduced in radioresistant cells. All in all, as GRHL3, helps LUSC cells escape from the immune surveillance and mediates radioresistance, it might be an attractive target for therapy-resistant LUSC.
Collapse
Affiliation(s)
- Haijun Wang
- Department of Thoracic Surgery, Xingtai People's Hospital, Xingtai 054000 Hebei, PR China
| | - Changjiang Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000 Hebei, PR China
| | - Chao Jiang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000 Hebei, PR China
| | - Yunjie Zhang
- Department of Surgical Oncology, Handan Central Hospital, Handan 056000 Hebei, PR China
| | - Xin Zhao
- School of Clinical Sciences, Hebei Medical University, Shijiazhuang 050000 Hebei, PR China
| | - Zhongfei Jia
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000 Hebei, PR China
| | - Jingchen Huo
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000 Hebei, PR China
| | - Jie Yang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000 Hebei, PR China.
| |
Collapse
|
12
|
Xing S, Chai H, Chen Z, Deng S, Nong F. Lycobetaine Has Therapeutic Efficacy in Lung Squamous Cell Carcinoma by Targeting USP32 to Trigger Ferroptosis. Curr Issues Mol Biol 2025; 47:163. [PMID: 40136417 PMCID: PMC11941616 DOI: 10.3390/cimb47030163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Ubiquitin-specific protease 32 (USP32), a deubiquitylating enzyme that controls the ubiquitin process, is overexpressed in multiple cancers and serves as a promising therapeutic target for cancer therapy. Drugs targeting ferroptosis have exhibited promising anticancer activity. Lycobetaine (LBT), a natural alkaloid, holds promise against various cancers, yet its specific targets and anticancer mechanisms remain unclear. In this study, we show that LBT induced ferroptosis in lung squamous cell carcinoma (LUSC) cells, accompanied by glutathione depletion and the accumulation of lipid peroxidation, malondialdehyde, and ferrous iron. Mechanistically, drug affinity responsive target stability-based mass spectrometry analysis, molecular dynamics simulations, and a cellular thermal shift assay confirmed that USP32 is a potential target of LBT in LUSC cells. Moreover, a strong interaction between USP32 and nuclear factor erythroid 2-related factor 2 (NRF2) was found via immunoprecipitation-mass spectrometry and co-immunoprecipitation. In addition, the ubiquitination assay results demonstrated that LBT treatment significantly increased NRF2 ubiquitination and degradation by targeting USP32. Importantly, USP32 overexpression effectively attenuated the effects of LBT on proliferation and ferroptosis in LUSC cells. In orthotopic LUSC xenografts, the administration of LBT significantly inhibited tumor growth and metastasis and induced ferroptosis by targeting the USP32-NRF2 signaling axis. Taken together, these data suggest that LBT exerts its anticancer effects by inhibiting USP32-mediated NRF2 deubiquitination to induce ferroptosis and that LBT may serve as a prospective USP32-targeting agent for LUSC treatment.
Collapse
Affiliation(s)
- Shangping Xing
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (S.X.); (H.C.); (Z.C.)
- Guangxi Key Laboratory for Bioactive Molecules Research and Evaluation, Nanning 530021, China
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Nanning 530021, China
| | - Hua Chai
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (S.X.); (H.C.); (Z.C.)
| | - Zhenlong Chen
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (S.X.); (H.C.); (Z.C.)
| | - Shuye Deng
- Department of Scientific Research, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530024, China
| | - Feifei Nong
- Department of Scientific Research, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530024, China
| |
Collapse
|
13
|
Liu Y, Kang L, Luo J, Yang M, Wang D, Ye J, Yang X, Wan W, Wong J, Xiao J. Targeting AKT as a promising strategy for SOX2-positive, chemoresistant osteosarcoma. Bone Res 2025; 13:25. [PMID: 39994220 PMCID: PMC11850766 DOI: 10.1038/s41413-024-00395-9] [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: 12/02/2023] [Revised: 11/06/2024] [Accepted: 12/01/2024] [Indexed: 02/26/2025] Open
Abstract
Osteosarcoma (OS) is the most prevalent type of primary malignant bone cancer and currently lacks effective targeted treatments. Increasing evidence indicates that SOX2 overexpression is a primary driver of OS. By screening a small-molecule kinase inhibitor library, we identified AKT as a kinase essential for robust SOX2 expression in OS cells. AKT was found to be frequently overexpressed in OS and positively correlated with SOX2 protein levels. We demonstrated that AKT has no effect on SOX2 transcription but promotes SOX2 protein stability. Mechanistically, AKT binds to and phosphorylates SOX2 at T116, preventing SOX2 ubiquitination and proteasome-dependent degradation by ubiquitin E3 ligases UBR5 and STUB1. Moreover, we found that AKT-SOX2 axis is a significant modulator of cancer stemness and chemoresistance and that the combination of AKT inhibitor MK2206 and cisplatin resulted in a synergistic and potent inhibition of OS tumor growth in the PDX model. In conclusion, we identified a critical role for AKT in promoting SOX2 overexpression, tumor stemness, and chemoresistance in OS, and provided evidence that targeting AKT combined with chemotherapy may hold promise for treating refractory OS. Working model showing that AKT stabilizes SOX2 by phosphorylating T116 site. Phosphorylation by AKT restraints the binding and ubiquitinoylation of SOX2 by the UBR5 and STUB1, thus promoting SOX2 stability and tumorigenic activity. Targeting AKT by MK2206 inhibits T116 phosphorylation and promotes SOX2 ubiquitination pathway, which impairs SOX2 tumorigenic activity. A combined treatment with chemo reagent and AKT inhibitor could achieve better therapeutic effect for SOX2-positive OS.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China
- Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Li Kang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jing Luo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Minglei Yang
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China
| | - Da Wang
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China
| | - Juelan Ye
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China
- Wuxi School of Medicine, Jiangnan University, Shanghai, China
| | - Xinghai Yang
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China
| | - Wei Wan
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China.
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jianru Xiao
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Navy Medical University, Shanghai, 200001, China.
| |
Collapse
|
14
|
Cheng Q, Leng D, Sun X, Feng J, Li F, Gao X, Wu T, Wei Q. Bi 2S 3/Bi 2O 2(OH) 2 nanorods with internal electric field throughout the entire bulk phase as photoelectrochemical sensing platforms for CYFRA21-1 immunoassay. Anal Chim Acta 2025; 1338:343591. [PMID: 39832861 DOI: 10.1016/j.aca.2024.343591] [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: 11/12/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
Photoelectrochemical (PEC) immunosensors are highly promising tools for monitoring biochemical molecules. Constructing high-performance heterojunctions is a general method to improve the sensitivity of PEC immunosensors. The internal electric field (IEF) formed at the heterojunction interface plays a crucial role in coordinating the separation of photogenerated carriers. Although IEF in heterojunctions shows extraordinary talents in PEC immunosensors, it is powerless to suppress the recombination of photogenerated carriers in the bulk phase. Thus, modulating the IEF of heterojunctions more than just at interfaces remains a challenge. Herein, a "signal-off" PEC immunosensor for CYFRA21-1 detection was constructed. The immunosensor was based on sulfurized bismuth oxyhydroxide compound Bi2S3/Bi2O2(OH)2 nanorods as the platforms, N-doped hollow homogenous Mo2C nanospheres with Ru NPs (Mo2C:N@Ru) as the quenching label. The Bi2O2(OH)2 were composed of [Bi2O2]2+ and OH- alternating layers, which provided an IEF throughout the bulk phase. Meanwhile, the heterogeneous interface further promoted the separation of photogenerated carriers and output strong photocurrent signals by energy level matching of Bi2O2(OH)2 with Bi2S3. Subsequently, Mo2C:N@Ru was quantitatively introduced in the presence of CYFRA21-1 through specific recognition between antigens and antibodies. The main structure of Mo2C:N@Ru was a homojunction formed by cubic and hexagonal phases Mo2C, which accelerated the separation of photogenerated carriers. Moreover, doped N element and Ru NPs further enhanced the competition between Mo2C:N@Ru and the Bi2S3/Bi2O2(OH)2 platforms for ascorbic acid and then achieved a quenching effect. The limit of detection for CYFRA21-1 was 17 fg/mL, and linearity range was 50 fg/mL -100 ng/mL. This PEC immunosensor achieves highly sensitive detection of CYFRA21-1 and has good scalability, which provides new ideas for the development analysis and detection platforms of tumor biomarkers. Simultaneously, this strategy might provide a novel perspective for modulating the distribution of the IEF of photoactive materials applied in PEC immunosensors.
Collapse
Affiliation(s)
- Qian Cheng
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China
| | - Dongquan Leng
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China
| | - Xu Sun
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China
| | - Jinhui Feng
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China
| | - Faying Li
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China
| | - Xing Gao
- College of Biological and Chemical Engineering, Qilu Institute of Technology, 250200, Jinan, PR China
| | - Tingting Wu
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China
| | - Qin Wei
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, PR China; Department of Chemistry, Sungkyunkwan University, 16419, Suwon, Republic of Korea.
| |
Collapse
|
15
|
Li Y, Li W, Liu Y, Peng Y, Tang J, Li X. Efficacy and safety of anlotinib combined with vinorelbine as second‑line treatment for elderly patients with advanced squamous cell lung carcinoma: A retrospective cohort. Mol Clin Oncol 2025; 22:21. [PMID: 39776939 PMCID: PMC11706458 DOI: 10.3892/mco.2024.2816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/06/2024] [Indexed: 01/05/2025] Open
Abstract
The aim of the present study was to investigate the efficacy and safety of anlotinib combined with vinorelbine (NVB) as a second-line treatment for elderly patients with advanced squamous cell lung carcinoma (SqCLC). The present retrospective analysis included 48 elderly patients (aged ≥65 years) diagnosed with advanced SqCLC who received anlotinib in combination with NVB as a second-line therapy between January 2021 and December 2023. The primary endpoints assessed were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR) and safety profile. The median PFS and OS for the cohort was found to be 5.0 and 9.5 months, respectively. By contrast, the ORR and DCR were found to be 29.17 and 70.83%. Further subgroup analysis indicated that patients who experienced specific adverse events (AEs), such as hypertension, proteinuria and hand-foot syndrome during treatment, generally had superior efficacy compared with those who did not experience these AEs (mPFS, 6.0 vs. 4.0 months; mOS, 11.0 vs. 8.5 months). In addition, apart from promising efficacy, patients who experienced common AEs also experienced decreased appetite (35.42%), fatigue (29.17%), hypertension (25%) and hand-foot syndrome (27.08%). Grade 3 or higher AEs occurred in <30% of patients, the majority of which was alleviated through corresponding support care. These results suggest that the combination of anlotinib and NVB as second-line therapy for elderly patients with advanced SqCLC demonstrated promising efficacy and a manageable safety profile. Such regimen may be a viable treatment option for this patient population. However, further prospective studies are required to validate these findings and optimize the dosing schedule for improved therapeutic outcomes.
Collapse
Affiliation(s)
- Yonghong Li
- The Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Wei Li
- The Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Yirui Liu
- Department of Nursing, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Yi Peng
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Jing Tang
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Xiaobing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
16
|
Long D, Ding Y, Wang P, Wei L, Ma K. Multi-Omics Analysis Reveals Immune Infiltration and Clinical Significance of Phosphorylation Modification Enzymes in Lung Adenocarcinoma. Int J Mol Sci 2025; 26:1066. [PMID: 39940833 PMCID: PMC11817228 DOI: 10.3390/ijms26031066] [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: 12/25/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Protein phosphorylation is a dynamic and reversible modification involved in almost all cellular processes. Numerous investigations have shown that protein phosphorylation modification enzymes (PPMEs) that regulate protein phosphorylation play an important role in the occurrence and treatment of tumors. However, there is still a lack of effective insights into the value of PPMEs in the classification and treatment of patients with lung adenocarcinoma (LUAD). Here, four topological algorithms identified 15 hub PPMEs from a protein-protein interaction (PPI) network. This PPI network was constructed using 124 PPMEs significantly correlated with 35 cancer hallmark-related pathways. Our study illustrates that these hub PPMEs can affect the survival of patients with LUAD in the form of somatic mutation or expression perturbation. Consistency clustering based on hub PPMEs recognized two phosphorylation modification subtypes (namely cluster1 and cluster2) from LUAD. Compared with patients in cluster1, the survival prognosis of patients in cluster2 is worse. This disparity is probably attributed to the higher tumor mutation burden, the higher male proportion, and the more significant expression disturbance in patients in cluster2. Moreover, phosphorylation modification subtypes also have different characteristics in terms of immune activity, immune infiltration level, immunotherapy response, and drug sensitivity. We constructed a PSig scoring system by using a principal component analysis algorithm to estimate the level of phosphorylation modification in individual LUAD patients. Patients in the high and low PSig score groups demonstrated different characteristics in terms of survival rate, tumor mutation burden, somatic gene mutation rate, immune cell abundance, and sensitivity to immunotherapy and drug treatment. This work reveals that phosphorylation plays a non-negligible role in the tumor microenvironment and immunotherapy of LUAD. Evaluating the phosphorylation status of individual LUAD patients by the PSig score can contribute to enhancing our cognition of the tumor microenvironment and guiding the formulation of more effective personalized treatment strategies.
Collapse
Affiliation(s)
- Deyu Long
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University Medical College, Shihezi 832000, China
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Yanheng Ding
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Lili Wei
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University Medical College, Shihezi 832000, China
| | - Ketao Ma
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University Medical College, Shihezi 832000, China
| |
Collapse
|
17
|
Yamaguchi F, Kondo C, Hirata K, Miyo K, Kanzaki M, Tei K, Kobayashi H. Efficacy of the PD-1 inhibitor penpulimab in combination with chemotherapy for advanced lung squamous cell carcinoma: insights from a phase III multicenter study. J Thorac Dis 2025; 17:10-14. [PMID: 39975732 PMCID: PMC11833557 DOI: 10.21037/jtd-24-1547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/12/2024] [Indexed: 02/21/2025]
Affiliation(s)
- Fumihiro Yamaguchi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Chika Kondo
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kento Hirata
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kenta Miyo
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Mamiko Kanzaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kazusawa Tei
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hitoshi Kobayashi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| |
Collapse
|
18
|
Jiang J, Huang LS. Special Issue "Molecular Advances and Perspectives of Lung Disease". Int J Mol Sci 2025; 26:946. [PMID: 39940714 PMCID: PMC11816814 DOI: 10.3390/ijms26030946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Respiratory diseases represent a significant global public health challenge, contributing to high mortality and morbidity rates worldwide [...].
Collapse
Affiliation(s)
- Jiacheng Jiang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China;
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Long Shuang Huang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China;
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
19
|
Dakal TC, Thakur M, George N, Singh TR, Yadav V, Kumar A. GTF2I acts as a novel tumor suppressor transcription factor and shows Favorable prognosis in renal cancer. Integr Biol (Camb) 2025; 17:zyaf001. [PMID: 39778513 DOI: 10.1093/intbio/zyaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/12/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
The role of GTF2I (General Transcription Factor2I) alteration has already been reported in thymic cancer as a valuable biomarker. However, the association of GTF2I mutation with renal cancer for prognosis of immunotherapy is not yet examined. The biologic and oncologic significance of GTF2I in renal cancer was examined at multiomics level such as mutation, copy number alteration, structural variants. The Cancer Genome Atlas (TCGA), Human Protein Atlas (HPA) were used to retrieve the omics data. The expression of GTF2I mRNA was quite significant in case of renal caner. Correlation among the GTF2I mRNA, mutation, CNA and structural variants was also studied. Interactome of GTF2I was also constructed using STRING database. Gain, amplification, and missense mutation exhibited a positive correlation between GTF2I mRNA expression and non-structural variants. Similarly, GTF2I mRNA expression and copy number alterations from GISTIC were positively correlated. High expression of GTF2I was associated with better overall survival indicating the less aggressive clinical features. Insight Box Investigating GTF2I's complex function as a tumor suppressor transcription factor in renal carcinoma provides fresh insights into its biologic and oncologic importance, especially when considering the prognosis of immunotherapy. Little is known about its possible use as a biomarker for renal cancer. Using a multiomics approach and utilizing information from the Human Protein Atlas (HPA) and The Cancer Genome Atlas (TCGA), our study clarifies the intricate relationship between mRNA expression, GTF2I changes, and clinical outcomes in renal cancer. Our results indicate that GTF2I expression may be used as a prognostic indicator because it is positively correlated with favorable survival outcomes. Furthermore, the molecular interactions behind GTF2I's functional significance in renal cancer are revealed by interactome analysis utilizing the STRING database, providing important information for further study and treatment approaches.
Collapse
MESH Headings
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/mortality
- Kidney Neoplasms/pathology
- Transcription Factors, TFII/metabolism
- Transcription Factors, TFII/genetics
- Prognosis
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Mutation
- DNA Copy Number Variations
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/mortality
- Carcinoma, Renal Cell/pathology
- Female
- Male
- Middle Aged
- Genes, Tumor Suppressor
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, University Road, Udaipur, Rajasthan 313001, India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Jant-Pali villages, Mahendergarh, Haryana 123031, India
| | - Nancy George
- Department of Biotechnology, Chandigarh University, NH-05 Chandigarh-Ludhiana Highway, Mohali, Punjab 140413, India
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan 173 234, H.P. India
| | - Vinod Yadav
- Department of Microbiology, Central University of Haryana, Jant-Pali villages, Mahendergarh, Haryana 123031, India
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India
| |
Collapse
|
20
|
Muneer G, Chen C, Chen Y. Advancements in Global Phosphoproteomics Profiling: Overcoming Challenges in Sensitivity and Quantification. Proteomics 2025; 25:e202400087. [PMID: 39696887 PMCID: PMC11735659 DOI: 10.1002/pmic.202400087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Protein phosphorylation introduces post-genomic diversity to proteins, which plays a crucial role in various cellular activities. Elucidation of system-wide signaling cascades requires high-performance tools for precise identification and quantification of dynamics of site-specific phosphorylation events. Recent advances in phosphoproteomic technologies have enabled the comprehensive mapping of the dynamic phosphoproteomic landscape, which has opened new avenues for exploring cell type-specific functional networks underlying cellular functions and clinical phenotypes. Here, we provide an overview of the basics and challenges of phosphoproteomics, as well as the technological evolution and current state-of-the-art global and quantitative phosphoproteomics methodologies. With a specific focus on highly sensitive platforms, we summarize recent trends and innovations in miniaturized sample preparation strategies for micro-to-nanoscale and single-cell profiling, data-independent acquisition mass spectrometry (DIA-MS) for enhanced coverage, and quantitative phosphoproteomic pipelines for deep mapping of cell and disease biology. Each aspect of phosphoproteomic analysis presents unique challenges and opportunities for improvement and innovation. We specifically highlight evolving phosphoproteomic technologies that enable deep profiling from low-input samples. Finally, we discuss the persistent challenges in phosphoproteomic technologies, including the feasibility of nanoscale and single-cell phosphoproteomics, as well as future outlooks for biomedical applications.
Collapse
Affiliation(s)
- Gul Muneer
- Institute of ChemistryAcademia SinicaTaipeiTaiwan
| | | | - Yu‐Ju Chen
- Institute of ChemistryAcademia SinicaTaipeiTaiwan
- Department of ChemistryNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
21
|
Nagano T, Takada K, Hashinokuchi A, Matsudo K, Kinoshita F, Akamine T, Kohno M, Shimokawa M, Takenaka T, Oda Y, Yoshizumi T. Clinical significance of CD155 expression in surgically resected lung squamous cell carcinoma. Int J Clin Oncol 2025; 30:62-71. [PMID: 39441454 DOI: 10.1007/s10147-024-02640-x] [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: 04/17/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Cluster of differentiation 155 (CD155) is expressed in many tumor types. CD155 is involved in the immune avoidance of tumor cells and contributes to tumor development and progression. Therefore, CD155 is a novel target for cancer immunotherapy. The clinical significance of CD155 expression in lung squamous cell carcinoma (LUSC) has not been fully elucidated. MATERIALS AND METHODS We performed a retrospective analysis of 264 patients with surgically resected LUSC. Immunohistochemistry was used to evaluate CD155 expression. The association of CD155 expression with clinicopathological features and clinical outcomes was assessed. We also analyzed the relationship between CD155 expression and programmed cell death-ligand 1 (PD-L1) expression and tumor-infiltrating lymphocytes. RESULTS Among the 264 patients, 137 patients (51.9%) were classified in the high CD155 expression group. High CD155 expression was significantly associated with pleural invasion, vascular invasion, PD-L1 positivity, and high CD3, CD4, and CD8 expressions. In multivariate analysis, the presence of pleural invasion and PD-L1 positivity were independent predictors of high CD155 expression. Kaplan-Meier curve analysis showed that high CD155 expression was significantly associated with shorter disease-free survival and overall survival. In multivariate analysis, high CD155 expression was an independent poor prognostic factor for overall survival, but not for disease-free survival. Subgroup analyses revealed that the prognostic effect of CD155 expression was observed in the PD-L1 positive group but not the PD-L1 negative group. CONCLUSION Our analysis revealed that high CD155 expression significantly predicted poor prognosis in patients with surgically resected LUSC, especially in patients with PD-L1-positive tumors.
Collapse
Affiliation(s)
- Taichi Nagano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Takada
- Department of Surgery, Saiseikai Fukuoka General Hospital, 1-3-46 Tenjin, Chuo-ku, Fukuoka, 810-0001, Japan.
| | - Asato Hashinokuchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoto Matsudo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumihiko Kinoshita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mikihiro Kohno
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
23
|
Wang X, Zhang C, Duan BJ, Bai J, Li T, Dong XY, Li EX. Forkhead box N1 is possibly a novel biomarker and prognostic indicator for patients with squamous cell lung carcinoma. Int J Med Sci 2024; 21:3058-3068. [PMID: 39628682 PMCID: PMC11610332 DOI: 10.7150/ijms.104616] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/02/2024] [Indexed: 12/06/2024] Open
Abstract
Background: The roles of Forkhead box N1 (FOXN1) in lung squamous cell carcinoma (LUSC) remains elusive. This study was focused on assessing the expression levels of FOXN1 in LUSC and exploring its potential clinical implications. Methods: Utilizing a range of databases, this study conducted an analysis of the FOXN1 gene's expression levels, comparing LUSC samples with those from normal lung tissues. The expression levels of FOXN1 in primary LUSC and corresponding normal lung tissues were assessed using immunohistochemistry (IHC). Histoscore was used to evaluate the staining degree. χ2 test and Fisher's exact test were employed to assess the association between categorical variables that do not possess an ordinal nature. Multivariate survival analysis was conducted using the Kaplan-Meier method, the Wilcoxon test, and the Cox proportional hazards model. Results: In contrast to normal lung tissues, the expression of the FOXN1 gene was found to be significantly elevated in LUSC tissues (P < 0.01). And FOXN1 was expressed in 79 (98.8%) evaluated LUSC tissues, most of which showed compositive IHC-staining intensity, presenting heterogeneously expression. 69 (87.3%) cases were characterized for strong immunostaining intensity, 70 (87.5%) cases showed moderate intensity, and 66 (82.5%) cases presented weak intensity. Only one sample of normal lung tissue, which represents 10% of the total, exhibited weak immunostaining exclusively (P < 0.05). Additionally, the expression of FOXN1 was found to have a significant correlation with the grading of LUSC, the presence of lymph node and distant metastases, the stage of the disease, and the survival outcomes (P < 0.05). Conclusion: The expression of FOXN1 is frequently increased in LUSC, and the patients with high FOXN1 expression have a poorer survival outcome. FOXN1 can be a novel biomarker and prognostic indicator for LUSC patients.
Collapse
Affiliation(s)
- Xi Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an,710061, China
| | - Caihua Zhang
- Department of Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Chongqing 404100, China
| | - Bao-Jun Duan
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an,710061, China
| | - Jun Bai
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an,710061, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xu-Yuan Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - En-Xiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
24
|
Wu J, Lu J, Chen Q, Chen H, Zheng Y, Cheng M. Pan-cancer analysis of CLDN18.2 shed new insights on the targeted therapy of upper gastrointestinal tract cancers. Front Pharmacol 2024; 15:1494131. [PMID: 39555091 PMCID: PMC11563798 DOI: 10.3389/fphar.2024.1494131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Background CLDN18.2 is a widely researched drug target. However, previous research has primarily been based on immunohistochemistry results and focused on gastric cancer. Methods To analyze the potential cancer-targeting effect of CLDN18.2 from a multi-omics perspective, this study quantified CLDN18.2 expression in The Cancer Genome Atlas (TCGA) pan-cancer cohort. Thus, the relationships between CLDN18.2 expression and genomic alterations, immune infiltration, and prognosis were analyzed. Additionally, we performed analyses of the differentially expressed genes and enriched pathways between the high- and low-CLDN18.2 expression groups, as well as the corresponding drug sensitivity analyses. Results The results indicated that CLDN18.2 was highly expressed in pancreatic adenocarcinoma (PAAD), stomach adenocarcinoma (STAD), colorectal cancer (CRC), and esophageal carcinoma (ESCA). Moreover, the high- and low-CLDN18.2 expression groups presented significant differences in terms of genomic alterations and immune infiltration, such as the levels of methylation and CD4+ T cell infiltration. Furthermore, high CLDN18.2 expression was significantly associated with poor prognosis in bladder urothelial carcinoma (BLCA), ESCA, and PAAD. In upper gastrointestinal tract cancers (STAD, ESCA, and PAAD), downregulated gene-enriched pathways were associated with cell signaling, whereas upregulated gene-enriched pathways were associated with angiogenesis. Finally, we identified drugs associated with CLDN18.2 expression to which samples with different levels of expression were differentially sensitive. Conclusion CLDN18.2 was highly expressed in upper gastrointestinal tract cancers, and its expression had a significant effect on genomic alterations and the tumor microenvironment. Additionally, low CLDN18.2 expression was linked to favorable prognosis. Our study reveals the potential value of CLDN18.2 for tumor prognosis and targeted therapy in various cancers, especially upper gastrointestinal tract cancers.
Collapse
Affiliation(s)
- Jun Wu
- Department of Clinical Laboratory, People's Hospital of Bao'an District, Shenzhen Baoan Clinical Medical College of Guangdong Medical University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinghua Lu
- Department of Clinical Laboratory, People's Hospital of Bao'an District, Shenzhen Baoan Clinical Medical College of Guangdong Medical University, Shenzhen, China
| | - Qiuyue Chen
- Department of Clinical Laboratory, People's Hospital of Bao'an District, Shenzhen Baoan Clinical Medical College of Guangdong Medical University, Shenzhen, China
| | - Haojie Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Minggang Cheng
- Department of Clinical Laboratory, People's Hospital of Bao'an District, Shenzhen Baoan Clinical Medical College of Guangdong Medical University, Shenzhen, China
| |
Collapse
|
25
|
Chen Q, Zheng X, Cheng W, Li J. Landscape of targeted therapies for lung squamous cell carcinoma. Front Oncol 2024; 14:1467898. [PMID: 39544292 PMCID: PMC11560903 DOI: 10.3389/fonc.2024.1467898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Lung cancer, a common type of malignant neoplasm, has seen significant advancements in the treatment of lung adenocarcinoma (LUAD). However, the management of lung squamous cell carcinoma (LSCC) continues to pose challenges. Traditional treatment methods for LSCC encompass surgical resection, chemotherapy, and radiotherapy. The introduction of targeted therapy and immunotherapy has greatly benefited LSCC patients, but issues such as limited immune response rates and adverse reactions persist. Therefore, gaining a deeper comprehension of the underlying mechanisms holds immense importance. This review provides an in-depth overview of classical signaling pathways and therapeutic targets, including the PI3K signaling pathway, CDK4/6 pathway, FGFR1 pathway and EGFR pathway. Additionally, we delve into alternative signaling pathways and potential targets that could offer new therapeutic avenues for LSCC. Lastly, we summarize the latest advancements in targeted therapy combined with immune checkpoint blockade (ICB) therapy for LSCC and discuss the prospects and challenges in this field.
Collapse
Affiliation(s)
- Qiuxuan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoshuo Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiting Cheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jian Li
- Institude of Experimental Immunology, University Clinic of Rheinische Friedrich-Wihelms-University, Bonn, Germany
| |
Collapse
|
26
|
Alexandru I, Davidescu L, Motofelea AC, Ciocarlie T, Motofelea N, Costachescu D, Marc MS, Suppini N, Șovrea AS, Coșeriu RL, Bondor DA, Bobeică LG, Crintea A. Emerging Nanomedicine Approaches in Targeted Lung Cancer Treatment. Int J Mol Sci 2024; 25:11235. [PMID: 39457017 PMCID: PMC11508987 DOI: 10.3390/ijms252011235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Lung cancer, the leading cause of cancer-related deaths worldwide, is characterized by its aggressive nature and poor prognosis. As traditional chemotherapy has the disadvantage of non-specificity, nanomedicine offers innovative approaches for targeted therapy, particularly through the development of nanoparticles that can deliver therapeutic agents directly to cancer cells, minimizing systemic toxicity and enhancing treatment efficacy. VEGF and VEGFR are shown to be responsible for activating different signaling cascades, which will ultimately enhance tumor development, angiogenesis, and metastasis. By inhibiting VEGF and VEGFR signaling pathways, these nanotherapeutics can effectively disrupt tumor angiogenesis and proliferation. This review highlights recent advancements in nanoparticle design, including lipid-based, polymeric, and inorganic nanoparticles, and their clinical implications in improving lung cancer outcomes, exploring the role of nanomedicine in lung cancer diagnoses and treatment.
Collapse
Affiliation(s)
- Isaic Alexandru
- Department X of General Surgery, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Lavinia Davidescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Tudor Ciocarlie
- Department VII Internal Medicine II, Discipline of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Nadica Motofelea
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
| | - Dan Costachescu
- Radiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Monica Steluta Marc
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Noemi Suppini
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Alina Simona Șovrea
- Department of Morphological Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Răzvan-Lucian Coșeriu
- Department of Microbiology, University of Medicine, Pharmacy, Science and Technology “George Emil Palade”, 540142 Târgu-Mures, Romania;
| | - Daniela-Andreea Bondor
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Laura-Gabriela Bobeică
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Andreea Crintea
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| |
Collapse
|
27
|
Wang L, Luo W, Zhang S, Zhang J, He L, Shi Y, Gao L, Wu B, Nie X, Hu C, Han X, He C, Xu B, Liang G. Macrophage-derived FGFR1 drives atherosclerosis through PLCγ-mediated activation of NF-κB inflammatory signalling pathway. Cardiovasc Res 2024; 120:1385-1399. [PMID: 38842387 DOI: 10.1093/cvr/cvae131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 06/07/2024] Open
Abstract
AIMS Atherosclerosis (AS) is a leading cause of cardiovascular morbidity and mortality. Atherosclerotic lesions show increased levels of proteins associated with the fibroblast growth factor receptor (FGFR) pathway. However, the functional significance and mechanisms governed by FGFR signalling in AS are not known. In the present study, we investigated fibroblast growth factor receptor 1 (FGFR1) signalling in AS development and progression. METHODS AND RESULTS Examination of human atherosclerotic lesions and aortas of Apoe-/- mice fed a high-fat diet (HFD) showed increased levels of FGFR1 in macrophages. We deleted myeloid-expressed Fgfr1 in Apoe-/- mice and showed that Fgfr1 deficiency reduces atherosclerotic lesions and lipid accumulations in both male and female mice upon HFD feeding. These protective effects of myeloid Fgfr1 deficiency were also observed when mice with intact FGFR1 were treated with FGFR inhibitor AZD4547. To understand the mechanistic basis of this protection, we harvested macrophages from mice and show that FGFR1 is required for macrophage inflammatory responses and uptake of oxidized LDL. RNA sequencing showed that FGFR1 activity is mediated through phospholipase-C-gamma (PLCγ) and the activation of nuclear factor-κB (NF-κB) but is independent of FGFR substrate 2. CONCLUSION Our study provides evidence of a new FGFR1-PLCγ-NF-κB axis in macrophages in inflammatory AS, supporting FGFR1 as a potentially therapeutic target for AS-related diseases.
Collapse
MESH Headings
- Animals
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Phospholipase C gamma/metabolism
- Phospholipase C gamma/genetics
- NF-kappa B/metabolism
- Signal Transduction
- Macrophages/metabolism
- Male
- Female
- Disease Models, Animal
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Aortic Diseases/immunology
- Humans
- Plaque, Atherosclerotic
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
- Lipoproteins, LDL/metabolism
- Diet, High-Fat
- Pyrazoles/pharmacology
- Inflammation Mediators/metabolism
- Benzamides/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Piperazines
Collapse
Affiliation(s)
- Lintao Wang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Wu Luo
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, The Affiliated First Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, Zhejiang 325035, China
| | - Suya Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Junsheng Zhang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
- Department of Pathology, Anhui Public Health Clinical Center, Hefei, Anhui 230032, China
| | - Lu He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Yifan Shi
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
| | - Li Gao
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Baochuan Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
| | - Xiaoyan Nie
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Chenghong Hu
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, The Affiliated First Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, Zhejiang 325035, China
| | - Xue Han
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
| | - Chaoyong He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, The Affiliated First Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
28
|
Chen X, Zhao W, Yu H, Wang S, Wang C, Song Y, Meng X, Li J. Case report: a case of lung squamous cell carcinoma with a novel FGFR3-IER5L fusion mutation responding to anlotinib. Front Oncol 2024; 14:1391349. [PMID: 39421453 PMCID: PMC11484447 DOI: 10.3389/fonc.2024.1391349] [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: 02/25/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024] Open
Abstract
Lung squamous cell carcinoma (LUSC) is the second most common pathological type of non-small cell lung cancer (NSCLC). However, compared with lung adenocarcinoma (LUAD), the incidence of driver gene mutations in LUSC is relatively lower and treatment options for LUSC patients are very limited. We described a LUSC patient with a novel FGFR3-IER5L fusion revealed by next generation sequencing in this report. The patient refused surgery, radiotherapy or chemotherapy and received anlotinib treatment. Anlotinib is a small molecular multi-target tyrosine kinase inhibitor, which can inhibit the activity of kinases including vascular endothelial growth factor receptor 2/3 (VEGFR2/3), fibroblast growth factor receptor 1-4 (FGFR1-4), platelet-derived growth factor receptor α/β (PDGFRα/β), and c-Kit. The patient achieved partial response and the progression-free survival was 3.8 months.
Collapse
Affiliation(s)
- Xiaoting Chen
- Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Wen Zhao
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hejiang Yu
- Department of Oncology, Yunyang County People’s Hospital, Chongqing, China
| | - Shuang Wang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chengjun Wang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yanan Song
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jisheng Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
29
|
Wang Y, Lei K, Zhao L, Zhang Y. Clinical glycoproteomics: methods and diseases. MedComm (Beijing) 2024; 5:e760. [PMID: 39372389 PMCID: PMC11450256 DOI: 10.1002/mco2.760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Glycoproteins, representing a significant proportion of posttranslational products, play pivotal roles in various biological processes, such as signal transduction and immune response. Abnormal glycosylation may lead to structural and functional changes of glycoprotein, which is closely related to the occurrence and development of various diseases. Consequently, exploring protein glycosylation can shed light on the mechanisms behind disease manifestation and pave the way for innovative diagnostic and therapeutic strategies. Nonetheless, the study of clinical glycoproteomics is fraught with challenges due to the low abundance and intricate structures of glycosylation. Recent advancements in mass spectrometry-based clinical glycoproteomics have improved our ability to identify abnormal glycoproteins in clinical samples. In this review, we aim to provide a comprehensive overview of the foundational principles and recent advancements in clinical glycoproteomic methodologies and applications. Furthermore, we discussed the typical characteristics, underlying functions, and mechanisms of glycoproteins in various diseases, such as brain diseases, cardiovascular diseases, cancers, kidney diseases, and metabolic diseases. Additionally, we highlighted potential avenues for future development in clinical glycoproteomics. These insights provided in this review will enhance the comprehension of clinical glycoproteomic methods and diseases and promote the elucidation of pathogenesis and the discovery of novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yujia Wang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Kaixin Lei
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Lijun Zhao
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Yong Zhang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
30
|
Li Q, Yang W, Zhang Q, Zhang D, Deng J, Chen B, Li P, Zhang H, Jiang Y, Li Y, Zhang B, Lin N. Wee1 inhibitor PD0166285 sensitized TP53 mutant lung squamous cell carcinoma to cisplatin via STAT1. Cancer Cell Int 2024; 24:315. [PMID: 39272147 PMCID: PMC11396119 DOI: 10.1186/s12935-024-03489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Lung squamous cell carcinoma (LUSCs) is associated with high mortality (20-30%) and lacks of effective treatments. Almost all LUSC exhibit somatic mutations in TP53. Wee1, a tyrosine kinase, regulates the cell cycle at the G2/M checkpoint. In TP53-deficient cells, the dependence on G2/M checkpoints increases. PD0166285 is the first reported drug with inhibitory activity against both Wee1 and PKMYT1. METHODS Protein expression was determined by Western blot analysis. Cell proliferation was assessed using cell colony formation and CCK-8 assays. Cell cycle was performed by PI staining with flow cytometry. Apoptosis was evaluated using Annexin V-Phycoerythrin double staining and flow cytometry. DNA damage was detected through comet assay and immunofluorescence assay. In vivo, apoptosis and anti-tumor effects were assessed using the TUNEL assay, a nude mouse model, and immunohistochemistry (IHC). Co-immunoprecipitation assay was used to detect protein-protein interactions. We analyzed Wee1, PKMYT1, and Stat1 expression in pan-cancer studies using the Ualcan public database and assessed their prognostic implications with Kaplan-Meier curves. RESULT PD0166285, a Wee1 inhibitor, effectively inhibits Wee1 activity, promoting cell entry into a mitotic crisis. Moreover, PD0166285 sensitizes cells to cisplatin, enhancing clinical outcomes. Our study demonstrated that PD016628 regulates the cell cycle through Rad51 and results in cell cycle arrest at the G2/M phase. We observed increased apoptosis in tumor cells treated with PD0166285, particularly when combined with cisplatin, indicating an enhanced apoptotic response. The upregulation of γ-H2AX serves as an indicator of mitotic catastrophe. Co-immunoprecipitation and data analysis revealed that apoptosis in LUSC is mediated through the Stat1 pathway, accompanied by decreased levels of Socs3. Furthermore, IHC staining confirmed significant differences in the expression of Phospho-CDK1 and γ-H2AX in LUSCs, suggesting involvement in DNA damage. CONCLUSIONS In summary, our study suggests that PD0166285, an inhibitor of Wee1, sensitizes LUSC cells to cisplatin and modulates DNA damage and apoptosis pathways through Rad51 and Stat1, respectively. These findings highlight the combination of PD0166285 and cisplatin as a promising therapeutic approach for treating LUSC.
Collapse
Affiliation(s)
- Qi Li
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China
| | - Wenjie Yang
- The Fourth Clinical College of Zhejiang, First People's Hospital, Chinese Medicine University, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Qingyi Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Daoming Zhang
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jun Deng
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Guangxi, 530021, China
| | - Binxin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China
| | - Ping Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China
| | - Huanqi Zhang
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yiming Jiang
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yangling Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Nengming Lin
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China.
| |
Collapse
|
31
|
Wei Y, Zhang R, Yin R, Wang S, Han J, Chen R, Fu Z. Immunotherapy Improves the Survival of Stage 4 Non-Small Cell Lung Cancer Patients at the US Population Level: The Real-World Evidence. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70000. [PMID: 39275901 PMCID: PMC11399776 DOI: 10.1111/crj.70000] [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: 02/27/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 09/16/2024]
Abstract
INTRODUCTION Immunotherapy has revolutionized the management of lung cancer and improved lung cancer survival in trials, but its real-world impact at the population level remains unclear. METHODS Using data obtained from eight Surveillance, Epidemiology, and End Results (SEER) registries from 2004 through 2019, we addressed the long-term trends in the incidence, incidence-based mortality (IBM), and survival of lung cancer patients in the United States. RESULTS The incidence and IBM of both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) all significantly decreased steadily from 2004 to 2019. The 1-year survival (1-YS) of both NSCLC and SCLC improved over time, with the best improvement observed for Stage 4 NSCLC. Two significant turning points of Stage 4 NSCLC 1-YS were observed over the years: 0.63% (95% confidence interval [CI]: 0.33%-0.93%) from 2004 to 2010, 0.81% (95% CI: 0.41%-1.21%) from 2010 to 2014 and a striking 2.09% (95% CI: 1.70%-2.47%) from 2014 to 2019. The same two turning points in 1-YS were pronounced for Stage 4 NSCLC in women, which were coincident with the introduction of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) and immunotherapy. However, for Stage 4 NSCLC in men, only one significant turning point in the 1-YS starting in 2014 was found, which might only correspond to immunotherapy. Significant period effects in reduced IBM were also observed for both Stage 4 AD and Stage 4 SQCC during the period. CONCLUSION This SEER analysis found that immunotherapy improved the survival of Stage 4 NSCLC patients at the population level in the United States. This real-world evidence confirms that immunotherapy has truly revolutionized the management of lung cancer.
Collapse
Affiliation(s)
- Yuxuan Wei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Zhang
- College of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Ruikang Yin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shijie Wang
- Radiation Oncology Department, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianglong Han
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenming Fu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
32
|
Liu T, Zhang E, Cui S, Dai H, Yang X, Lin C. Effects of 630 nm laser on apoptosis, metastasis, invasion and epithelial-to-mesenchymal transition of human lung squamous cell carcinoma H520 cells mediated by hematoporphyrin derivatives. Lasers Med Sci 2024; 39:228. [PMID: 39210165 DOI: 10.1007/s10103-024-04176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Photodynamic therapy (PDT) has significant advantages in the treatment of malignant lung tumors. The research on the mechanism of PDT mediated by hematoporphyrin derivatives (HPD) and its cytotoxic effects on lung cancer cells has primarily focused on lung adenocarcinoma cells. However, the impact of HPD-PDT on lung squamous cell carcinoma has not been thoroughly studied. This study aimed to investigate the effects of 630 nm laser on apoptosis, metastasis, invasion, and epithelial-mesenchymal transition (EMT) in human lung squamous cell carcinoma H520 cells mediated by HPD. H520 cells were divided into four groups: control group, photosensitizer group, irradiation group, and HPD-PDT group. Cell proliferation was assessed using CCK8 assay; cell apoptosis was detected by Hoechst 33258 staining and flow cytometry; cell migration and invasion abilities were evaluated using wound-healing and invasion assays; and protein and mRNA expressions were analyzed by Western blot and reverse transcription-polymerase chain reaction (RT-PCR) respectively. Results showed that HPD-PDT significantly inhibited cell proliferation, promoted apoptosis (P < 0.05), suppressed cell migration and invasion (P < 0.05), decreased Bcl-2 mRNA expression, and increased Bax and Caspase-9 mRNA expression(P < 0.05). Western blotting analysis indicated increased expression of Bax, Caspase-9, and E-cadherin, and decreased expression of Bcl-2, N-cadherin, and Vimentin (P < 0.05). In conclusion, 630 nm laser mediated by HPD promoted cell apoptosis via upregulation of Bax and caspase-9, and downregulation of Bcl-2, and inhibited cell migration and invasion by regulating EMT in H520 cells.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Enhua Zhang
- Department of Respiratory and Critical Care Medicine, Linyi Central Hospital, Linyi, China
| | - Shichao Cui
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haoyu Dai
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaohui Yang
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cunzhi Lin
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
33
|
Li M, Chen L, Yu F, Mei H, Ma X, Ding K, Yang Y, Rong Z. CTDSPL2 promotes the progression of non-small lung cancer through PI3K/AKT signaling via JAK1. Cell Death Discov 2024; 10:389. [PMID: 39209829 PMCID: PMC11362329 DOI: 10.1038/s41420-024-02162-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Carboxy-terminal domain small phosphatase like 2 (CTDSPL2), one of the haloacid dehalogenase phosphatases, is associated with several diseases including cancer. However, the role of CTDSPL2 and its regulatory mechanism in lung cancer remain unclear. Here, we aimed to explore the clinical implications, biological functions, and molecular mechanisms of CTDSPL2 in non-small cell lung cancer (NSCLC). CTDSPL2 was identified as a novel target of the tumor suppressor miR-193a-3p. CTDSPL2 expression was significantly elevated in NSCLC tissues. Database analysis showed that CTDSPL2 expression was negatively correlated with patient survival. Depletion of CTDSPL2 inhibited the proliferation, migration, and invasion of NSCLC cells, as well as tumor growth and metastasis in mouse models. Additionally, silencing of CTDSPL2 enhanced CD4+ T cell infiltration into tumors. Moreover, CTDSPL2 interacted with JAK1 and positively regulated JAK1 expression. Subsequent experiments indicated that CTDSPL2 activated the PI3K/AKT signaling pathway through the upregulation of JAK1, thereby promoting the progression of NSCLC. In conclusion, CTDSPL2 may play an oncogenic role in NSCLC progression by activating PI3K/AKT signaling via JAK1. These findings may provide a potential target for the diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Muzi Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - La Chen
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Fangfang Yu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Huijuan Mei
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xingxing Ma
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, 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
| | - Yanan Yang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| | - Ziye Rong
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
34
|
Ding Z, Chen J, Li B, Ji X. Inflammatory factors and risk of lung adenocarcinoma: a Mendelian randomization study mediated by blood metabolites. Front Endocrinol (Lausanne) 2024; 15:1446863. [PMID: 39257908 PMCID: PMC11384989 DOI: 10.3389/fendo.2024.1446863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/29/2024] [Indexed: 09/12/2024] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common type of lung cancer, and its pathogenesis remains not fully elucidated. Inflammation and metabolic dysregulation are considered to play crucial roles in LUAD development, but their causal relationships and specific mechanisms remain unclear. Methods This study employed a two-sample Mendelian randomization (MR) approach to systematically evaluate the causal associations between 91 circulating inflammatory factors, 1,400 serum metabolites, and LUAD. We utilized LUAD genome-wide association studies (GWAS) data from the FinnGen biobank and GWAS data of metabolites and inflammatory factors from the GWAS catalog to conduct two-sample MR analyses. For the identified key metabolites, we further used mediator MR to investigate their mediating effects in the influence of IL-17A on LUAD and explored potential mechanisms through protein-protein interaction and functional enrichment analyses. Results The MR analyses revealed that IL-17A (OR 0.78, 95%CI 0.62-0.99) was negatively associated with LUAD, while 71 metabolites were significantly associated with LUAD. Among them, ferulic acid 4-sulfate may play a crucial mediating role in the suppression of LUAD by IL-17A (OR 0.87, 95%CI 0.78-0.97). IL-17A may exert its anti-LUAD effects through extensive interactions with genes related to ferulic acid 4-sulfate metabolism (such as SULT1A1, CYP1A1, etc.), inhibiting oxidative stress and inflammatory responses, as well as downstream tumor-related pathways of ferulic acid 4-sulfate (such as MAPK, NF-κB, etc.). Conclusion This study discovered causal associations between IL-17A, multiple serum metabolites, and LUAD occurrence, revealing the key role of inflammatory and metabolic dysregulation in LUAD pathogenesis. Our findings provide new evidence-based medical support for specific inflammatory factors and metabolites as early predictive and risk assessment biomarkers for LUAD, offering important clues for subsequent mechanistic studies and precision medicine applications.
Collapse
Affiliation(s)
- Zheng Ding
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| | - Juan Chen
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| | - Bohan Li
- Department of Urinary Surgery, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| | - Xinyu Ji
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| |
Collapse
|
35
|
Zhong H, Wang L, Zhu X, Li S, Li X, Ding C, Wang K, Wang X. STAT3 inhibitor Stattic Exhibits the Synergistic Effect with FGFRs Inhibitor Erdafitinib in FGFR1-positive Lung Squamous Cell Carcinoma. J Cancer 2024; 15:5415-5424. [PMID: 39247610 PMCID: PMC11375536 DOI: 10.7150/jca.97477] [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: 04/18/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Lung squamous cell carcinoma (LUSC), a subset of non-small cell lung cancer (NSCLC), accounts for about 30% of all lung cancers (LC) and exhibits a dismal response to current therapeutic protocols. Existed studies have indicated that aberrations in fibroblast growth factor receptors (FGFRs) play a pivotal role in the progression of LUSC, rendering them as attractive targets for therapeutic intervention in this cancer type. This study found that Erdafitinib (Erda), a novel pan-FGF receptor tyrosine kinase inhibitor (TKI), exerted a cytotoxic effect on LUSC cells. However, STAT3, the downstream target of FGFRs, remained still activated despite Erdafitinib treatment. Then, a STAT3 inhibitor, Stattic (Sta), was concurrently used with Erdafitinib, and the combined treatment demonstrated a synergistic efficacy in both in vitro and in vivo models of LUSC when compared to that of the treatment of the Erdafitinib or Stattic alone. Further molecular studies showed that such an effect of Erdafitinib and Stattic was associated with their concurrently inhibitory effect on FGFR1 and STAT3 signaling in LUSC cells. Therefore, the findings of this study indicated that the concurrent use of Erdafitinib and Stattic is a promising therapeutic approach for the treatment of FGFR1-positive LUSC.
Collapse
Affiliation(s)
- Hongqin Zhong
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
- Nantong University Medical School, Nantong 226007, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Shu Li
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214126, Jiangsu Province, China
| | - Xiyue Li
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
| | - Chen Ding
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Xun Wang
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
- Nantong University Medical School, Nantong 226007, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214126, Jiangsu Province, China
| |
Collapse
|
36
|
Zhang D, Li J, Zhang C, Xue J, Li P, Shang K, Zhang X, Lang B. The deubiquitinating enzyme USP35 regulates the stability of NRF2 protein. Open Life Sci 2024; 19:20220935. [PMID: 39156988 PMCID: PMC11330172 DOI: 10.1515/biol-2022-0935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
Many cancers exhibit resistance to chemotherapy, resulting in a poor prognosis. The transcription factor NRF2, activated in response to cellular antioxidants, plays a crucial role in cell survival, proliferation, and resistance to chemotherapy. This factor may serve as a promising target for therapeutic interventions in esophageal carcinoma. Recent research suggests that NRF2 activity is modulated by ubiquitination mediated by the KEAP1-CUL3 E3 ligase complex, highlighting the importance of deubiquitination. However, the specific deubiquitinase responsible for regulating NRF2 in esophageal cancer remains unknown. In this study, a novel regulator of the NRF2 protein, Ubiquitin-Specific Protease 35 (USP35), has been identified. Mechanistically, USP35 modulates NRF2 stability through enzymatic deubiquitination. USP35 interacts with NRF2 and facilitates its deubiquitination. Knockdown of USP35 leads to a notable increase in NRF2 levels and enhances the sensitivity of cells to chemotherapy. These findings suggest that the USP35-NRF2 axis is a key player in the regulation of therapeutic strategies for esophageal cancer.
Collapse
Affiliation(s)
- Dian Zhang
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Jiawen Li
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Chao Zhang
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Jinliang Xue
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Peihao Li
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Kai Shang
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Xiao Zhang
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| | - Baoping Lang
- Department of Thoracic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Xigong District, Luoyang, China
| |
Collapse
|
37
|
Deng R, Zhang W, Lv J, Wang F, Chen Y, Jiang C, Guan Y, Zhang C. Afatinib as first-line treatment for advanced lung squamous cell carcinoma harboring uncommon EGFR G719C and S768I co-mutation: A case report and literature review. Heliyon 2024; 10:e35304. [PMID: 39166093 PMCID: PMC11334663 DOI: 10.1016/j.heliyon.2024.e35304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Ten percent of non-small cell lung cancer patients with epidermal growth factor receptor (EGFR) mutations harbor uncommon variants. These mutations are mainly involved in lung adenocarcinomas but are rare in lung squamous cell carcinoma (LSCC). In 2018, the Food and Drug Administration-approved afatinib for this specific patient population. However, there is limited information regarding the effectiveness of afatinib for LSCC with EGFR mutations. This case report documented a unique case of a patient with LSCC, which had a rare compound EGFR mutation (G719C and S768I) and showed significant response to afatinib treatment, with 10 months of progression-free survival. New NTRK1 and RET gene mutations may play a potential role in the development of acquired resistance to afatinib following clinical progression. This case highlights the importance of genetic profiling in patients with LSCC. Although these patients have a low positive rate of EGFR mutations, searching for EGFR mutations in these patients might broaden their treatment options.
Collapse
Affiliation(s)
- Ruoyu Deng
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Wen Zhang
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Jialing Lv
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Fang Wang
- Department of Pathology, Second People's Hospital of Qujing City, Qujing, 655000, China
| | - Yanqiong Chen
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Chengqi Jiang
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Yaling Guan
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Chao Zhang
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| |
Collapse
|
38
|
Wu W, Chen Z, Wen H, Zhang H. Unveiling potential drug targets for lung squamous cell carcinoma through the integration of druggable genome and genome-wide association data. Front Genet 2024; 15:1431684. [PMID: 39175755 PMCID: PMC11338847 DOI: 10.3389/fgene.2024.1431684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Background: Lung squamous cell carcinoma (LSCC) is a major subtype of lung cancer with poor prognosis and low survival rate. Compared with lung adenocarcinoma, yet no FDA-approved targeted-therapy has been found for lung squamous cell carcinoma. Methods: To identify potential drug targets for LSCC, Summary-data-based Mendelian randomization (SMR) analysis was used to examine the potential association between 4,543 druggable genes and LSCC, followed by colocalization analysis and HEIDI tests to confirm the robustness of the result. Phenome-wide association study (PheWAS) explored potential side effects of candidate drug targets. Enrichment analysis and protein-protein interaction networks revealed the function and significance of therapeutic targets. Single-cell expression analysis was used to examine cell types with enrichment expression of druggable genes in LSCC tissue. Drug prediction included screening potential drug candidates and evaluating their interactions with targets through molecular docking. Results: This research has identified ten significant drug targets for LSCC through a comprehensive SMR analysis. These targets included (COPA, PKD2L1, CCR1, C2, CYP21A2, and NCSTN as risk factors, and CCNA2, C4A, APOM, and LPAR2 as protective factors). PheWAS demonstrated that C2, CCNA2, LPAR2, and NCSTN exhibited associations with other phenotypes at the genetic level. Then, we found four potentially effective drugs with the Dsigdb database. Subsequently, molecular docking indicated that favorable binding interactions between drug candidates and potential target molecules. In the druggability evaluation, five out of ten drug target genes have been used in drug development (APOM, C4A, CCNA2, COPA, and PKD2L1). Six out of ten druggable genes showed significant expression in LSCC tissues (COPA, PKD2L1, CCR1, C2, NCSTN, LPAR2). Besides, Single-cell expression analysis revealed that C2 and CCNA2 were primarily enriched in macrophages, while COPA and NCSTN were enriched in both macrophages and epithelial cells. Conclusion: Our research revealed ten potential druggable genes for LSCC treatment, which might help to advance the precise and efficient therapeutic approaches of LSCC.
Collapse
Affiliation(s)
- Wenhua Wu
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhengrui Chen
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haiteng Wen
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyun Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hosptial, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Whitehead CE, Ziemke EK, Frankowski-McGregor CL, Mumby RA, Chung J, Li J, Osher N, Coker O, Baladandayuthapani V, Kopetz S, Sebolt-Leopold JS. A first-in-class selective inhibitor of EGFR and PI3K offers a single-molecule approach to targeting adaptive resistance. NATURE CANCER 2024; 5:1250-1266. [PMID: 38992135 PMCID: PMC11357990 DOI: 10.1038/s43018-024-00781-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/09/2024] [Indexed: 07/13/2024]
Abstract
Despite tremendous progress in precision oncology, adaptive resistance mechanisms limit the long-term effectiveness of molecularly targeted agents. Here we evaluated the pharmacological profile of MTX-531 that was computationally designed to selectively target two key resistance drivers, epidermal growth factor receptor and phosphatidylinositol 3-OH kinase (PI3K). MTX-531 exhibits low-nanomolar potency against both targets with a high degree of specificity predicted by cocrystal structural analyses. MTX-531 monotherapy uniformly resulted in tumor regressions of squamous head and neck patient-derived xenograft (PDX) models. The combination of MTX-531 with mitogen-activated protein kinase kinase or KRAS-G12C inhibitors led to durable regressions of BRAF-mutant or KRAS-mutant colorectal cancer PDX models, resulting in striking increases in median survival. MTX-531 is exceptionally well tolerated in mice and uniquely does not lead to the hyperglycemia commonly seen with PI3K inhibitors. Here, we show that MTX-531 acts as a weak agonist of peroxisome proliferator-activated receptor-γ, an attribute that likely mitigates hyperglycemia induced by PI3K inhibition. This unique feature of MTX-531 confers a favorable therapeutic index not typically seen with PI3K inhibitors.
Collapse
Affiliation(s)
- Christopher E Whitehead
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- MEKanistic Therapeutics, Inc., Ann Arbor, MI, USA
| | | | | | - Rachel A Mumby
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - June Chung
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Jinju Li
- Department of Biostatistics, The University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Nathaniel Osher
- Department of Biostatistics, The University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Oluwadara Coker
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Veerabhadran Baladandayuthapani
- Department of Biostatistics, The University of Michigan School of Public Health, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Judith S Sebolt-Leopold
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA.
- MEKanistic Therapeutics, Inc., Ann Arbor, MI, USA.
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA.
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
40
|
Shi K, Shen C, Xie Y, Fu L, Zhang S, Wang K, Naeem S, Yuan Z. Prognostic predictive modeling of non-small cell lung cancer associated with cadmium-related pathogenic genes. Comput Biol Chem 2024; 111:108096. [PMID: 38788566 DOI: 10.1016/j.compbiolchem.2024.108096] [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: 02/06/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Persistent exposure to low-dose of cadmium is strongly linked to both the development and prognosis of non-small cell lung cancer (NSCLC), yet the precise molecular mechanism behind this relationship remains uncertain. In this study, cadmium-related pathogenic genes (CRPGs) in NSCLC were identified via differential expression analysis. NSCLC patient clusters related to CRPGs were constructed through univariate Cox and K-means clustering algorithms. Multivariate Cox and least absolute shrinkage and selection operator (LASSO) regression analyses were employed to determine the prognosis. Sixteen CRPGs showed a significant association with NSCLC. We found biological and prognostic differences between patients in clusters A and B. A predictive prognostic risk model for NSCLC revealed that FAM83H, MSMO1, and SNAI1 are central. Hence, the 3 hub genes were named. To further elucidate the role of CRPGs in NSCLC, A549 cells were exposed to CdCl2. The mRNA and protein expression levels of the 3 hub genes and cell invasion were detected. Moreover, 10 μM CdCl2 may increase the protein expression of 3 hub genes and enhance the invasive ability of A549 cells. This risk model may have established a theoretical foundation for investigating the mechanisms, treatment, and prognosis of NSCLC.
Collapse
Affiliation(s)
- Kejian Shi
- School of Public Health, Wuhan University, 115 Donghu Road, Wuhan, Hubei Province 430071, China
| | - Chao Shen
- College of Life Sciences, Wuhan University, 299 Bayi Road, Wuhan, Hubei Province 430072, China
| | - Yaxuan Xie
- Wuhan Children's Hospital, Tongji Medical College Huazhong University of Science and Technology, 100 Xianggang Road, Wuhan, Hubei Province 430010, China
| | - Liangying Fu
- School of Public Health, Wuhan University, 115 Donghu Road, Wuhan, Hubei Province 430071, China
| | - Shihan Zhang
- Xuchang Vocational Technical College, 4336 Xinxing Road, Xuchang, Henan Province 461000, China
| | - Kai Wang
- The First Hospital of Wuhan City, 215 Zhongshan Avenue, Wuhan, Hubei Province 430022, China
| | - Shafaq Naeem
- School of Public Health, Wuhan University, 115 Donghu Road, Wuhan, Hubei Province 430071, China
| | - Zhanpeng Yuan
- School of Public Health, Wuhan University, 115 Donghu Road, Wuhan, Hubei Province 430071, China.
| |
Collapse
|
41
|
Kong Y, Jiang R, Zhou H, Ge M, Lin H, Wang Y, Yao R, Wang Q, Liang X, Li J, Zhou X. PHF12 regulates HDAC1 to promote tumorigenesis via EGFR/AKT signaling pathway in non-small cell lung cancer. J Transl Med 2024; 22:689. [PMID: 39075515 PMCID: PMC11287983 DOI: 10.1186/s12967-024-05488-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Lung cancer stands as the second most prevalent malignant neoplasm worldwide. Addressing the underlying mechanisms propelling the progression of non-small cell lung cancer is of paramount importance. In this study, we have elucidated the pivotal role of PHF12 in this context. MATERIALS AND METHODS We harnessed clinical lung cancer tissue samples and non-small cell lung cancer cell lines to discern the expression pattern of PHF12. In vitro assays probing cell proliferation were conducted to substantiate the functional impact of PHF12. Furthermore, an in vivo Xenograft model was employed to dissect the role of PHF12. Employing ChIP assays and qRT-PCR, we delved into the intricate binding dynamics between PHF12 and HDAC1. Mechanistic insights into the PHF12-HDAC1 axis in lung cancer progression were pursued via RNA-seq and GSEA analyses. RESULTS Notably, PHF12 exhibited a substantial upregulation within tumor tissue, concomitant with its correlation to HDAC1. The trilogy of cell proliferation assays, transwell assays, and the Xenograft model collectively underscored the promoting influence of PHF12 on lung cancer proliferation, both in vitro and in vivo. The ChIP assay unveiled the transcriptional regulatory role of PHF12 in governing HDAC1 expression. This correlation extended to both mRNA and protein levels. PHF12 promotes NSCLC progression through regulating HDCA1 expression. Intriguingly, the rescue of function within NSCLC cell lines post PHF12 knockdown was achievable through HDAC1 overexpression. Additionally, our findings unveiled the capacity of the PHF12-HDAC1 axis to activate the EGFR/AKT signaling pathway, thereby further corroborating its significance in lung cancer progression. CONCLUSION Our study identified PHF12 as an oncogenic role in lung cancer proliferation and migration for the first time. PHF12 transcriptionally regulate HDAC1 and activate EGFR/AKT signaling pathway in NSCLC progression. PHF12 may serve as an important target in lung cancer therapy.
Collapse
Affiliation(s)
- Yiru Kong
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rongrong Jiang
- Department of Cardiothoracic Surgery, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Hui Zhou
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Mengxi Ge
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Hao Lin
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Yu Wang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Rongrong Yao
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Qing Wang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Xiaohua Liang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Jing Li
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China.
| | - Xinli Zhou
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China.
| |
Collapse
|
42
|
Chen H, Xu Y, Lin H, Wan S, Luo L. A prognostic framework for predicting lung signet ring cell carcinoma via a machine learning based cox proportional hazard model. J Cancer Res Clin Oncol 2024; 150:364. [PMID: 39052087 PMCID: PMC11272739 DOI: 10.1007/s00432-024-05886-0] [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: 08/05/2023] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE Signet ring cell carcinoma (SRCC) is a rare type of lung cancer. The conventional survival nomogram used to predict lung cancer performs poorly for SRCC. Therefore, a novel nomogram specifically for studying SRCC is highly required. METHODS Baseline characteristics of lung signet ring cell carcinoma were obtained from the Surveillance, Epidemiology, and End Results (SEER) database. Univariate and multivariate Cox regression and random forest analysis were performed on the training group data, respectively. Subsequently, we compared results from these two types of analyses. A nomogram model was developed to predict 1-year, 3-year, and 5-year overall survival (OS) for patients, and receiver operating characteristic (ROC) curves and calibration curves were used to assess the prediction accuracy. Decision curve analysis (DCA) was used to assess the clinical applicability of the proposed model. For treatment modalities, Kaplan-Meier curves were adopted to analyze condition-specific effects. RESULTS We obtained 731 patients diagnosed with lung signet ring cell carcinoma (LSRCC) in the SEER database and randomized the patients into a training group (551) and a validation group (220) with a ratio of 7:3. Eight factors including age, primary site, T, N, and M.Stage, surgery, chemotherapy, and radiation were included in the nomogram analysis. Results suggested that treatment methods (like surgery, chemotherapy, and radiation) and T-Stage factors had significant prognostic effects. The results of ROC curves, calibration curves, and DCA in the training and validation groups demonstrated that the nomogram we constructed could precisely predict survival and prognosis in LSRCC patients. Through deep verification, we found the constructed model had a high C-index, indicating that the model had a strong predictive power. Further, we found that all surgical interventions had good effects on OS and cancer-specific survival (CSS). The survival curves showed a relatively favorable prognosis for T0 patients overall, regardless of the treatment modality. CONCLUSIONS Our nomogram is demonstrated to be clinically beneficial for the prognosis of LSRCC patients. The surgical intervention was successful regardless of the tumor stage, and the Cox proportional hazard (CPH) model had better performance than the machine learning model in terms of effectiveness.
Collapse
Affiliation(s)
- Haixin Chen
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Yanyan Xu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Haowen Lin
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Shibiao Wan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
43
|
Zhang L, Xu LY, Tang F, Liu D, Zhao XL, Zhang JN, Xia J, Wu JJ, Yang Y, Peng C, Ao H. New perspectives on the therapeutic potential of quercetin in non-communicable diseases: Targeting Nrf2 to counteract oxidative stress and inflammation. J Pharm Anal 2024; 14:100930. [PMID: 39005843 PMCID: PMC11245930 DOI: 10.1016/j.jpha.2023.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 07/16/2024] Open
Abstract
Non-communicable diseases (NCDs), including cardiovascular diseases, cancer, metabolic diseases, and skeletal diseases, pose significant challenges to public health worldwide. The complex pathogenesis of these diseases is closely linked to oxidative stress and inflammatory damage. Nuclear factor erythroid 2-related factor 2 (Nrf2), a critical transcription factor, plays an important role in regulating antioxidant and anti-inflammatory responses to protect the cells from oxidative damage and inflammation-mediated injury. Therefore, Nrf2-targeting therapies hold promise for preventing and treating NCDs. Quercetin (Que) is a widely available flavonoid that has significant antioxidant and anti-inflammatory properties. It modulates the Nrf2 signaling pathway to ameliorate oxidative stress and inflammation. Que modulates mitochondrial function, apoptosis, autophagy, and cell damage biomarkers to regulate oxidative stress and inflammation, highlighting its efficacy as a therapeutic agent against NCDs. Here, we discussed, for the first time, the close association between NCD pathogenesis and the Nrf2 signaling pathway, involved in neurodegenerative diseases (NDDs), cardiovascular disease, cancers, organ damage, and bone damage. Furthermore, we reviewed the availability, pharmacokinetics, pharmaceutics, and therapeutic applications of Que in treating NCDs. In addition, we focused on the challenges and prospects for its clinical use. Que represents a promising candidate for the treatment of NCDs due to its Nrf2-targeting properties.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jia Xia
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jiao-Jiao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hui Ao
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
44
|
Fabrizio FP, Sparaneo A, Gorgoglione G, Battista P, Centra F, Delli Muti F, Trombetta D, Centonza A, Graziano P, Rossi A, Fazio VM, Muscarella LA. Effects of KEAP1 Silencing on NRF2 and NOTCH Pathways in SCLC Cell Lines. Cancers (Basel) 2024; 16:1885. [PMID: 38791966 PMCID: PMC11120002 DOI: 10.3390/cancers16101885] [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: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
The KEAP1/NRF2 pathway is a master regulator of several redox-sensitive genes implicated in the resistance of tumor cells against therapeutic drugs. The dysfunction of the KEAP1/NRF2 system has been correlated with neoplastic patients' outcomes and responses to conventional therapies. In lung tumors, the growth and the progression of cancer cells may also involve the intersection between the molecular NRF2/KEAP1 axis and other pathways, including NOTCH, with implications for antioxidant protection, survival of cancer cells, and drug resistance to therapies. At present, the data concerning the mechanism of aberrant NRF2/NOTCH crosstalk as well as its genetic and epigenetic basis in SCLC are incomplete. To better clarify this point and elucidate the contribution of NRF2/NOTCH crosstalk deregulation in tumorigenesis of SCLC, we investigated genetic and epigenetic dysfunctions of the KEAP1 gene in a subset of SCLC cell lines. Moreover, we assessed its impact on SCLC cells' response to conventional chemotherapies (etoposide, cisplatin, and their combination) and NOTCH inhibitor treatments using DAPT, a γ-secretase inhibitor (GSI). We demonstrated that the KEAP1/NRF2 axis is epigenetically controlled in SCLC cell lines and that silencing of KEAP1 by siRNA induced the upregulation of NRF2 with a consequent increase in SCLC cells' chemoresistance under cisplatin and etoposide treatment. Moreover, KEAP1 modulation also interfered with NOTCH1, HES1, and DLL3 transcription. Our preliminary data provide new insights about the downstream effects of KEAP1 dysfunction on NRF2 and NOTCH deregulation in this type of tumor and corroborate the hypothesis of a cooperation of these two pathways in the tumorigenesis of SCLC.
Collapse
Affiliation(s)
- Federico Pio Fabrizio
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Angelo Sparaneo
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| | - Giusy Gorgoglione
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| | - Pierpaolo Battista
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| | - Flavia Centra
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| | - Francesco Delli Muti
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| | - Domenico Trombetta
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| | - Antonella Centonza
- Oncology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Paolo Graziano
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Antonio Rossi
- Oncology Center of Excellence, Therapeutic Science & Strategy Unit, IQVIA, 20124 Milan, Italy
| | - Vito Michele Fazio
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
- Department of Medicine, Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128 Rome, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00185 Rome, Italy
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.S.); (G.G.); (P.B.); (F.C.); (F.D.M.); (D.T.); (V.M.F.)
| |
Collapse
|
45
|
Liu D, Zuo R, Liu W, He Y, Wang Y, Yue P, Gong W, Cui J, Zhu F, Luo Y, Qi L, Guo Y, Chen L, Li G, Liu Z, Chen P, Guo H. DNAJC24 acts directly with PCNA and promotes malignant progression of LUAD by activating phosphorylation of AKT. FASEB J 2024; 38:e23630. [PMID: 38713100 DOI: 10.1096/fj.202300667rr] [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: 04/07/2023] [Revised: 03/09/2024] [Accepted: 04/12/2024] [Indexed: 05/08/2024]
Abstract
Heat shock proteins (HSPs) are a group of highly conserved proteins found in a wide range of organisms. In recent years, members of the HSP family were overexpressed in various tumors and widely involved in oncogenesis, tumor development, and therapeutic resistance. In our previous study, DNAJC24, a member of the DNAJ/HSP40 family of HSPs, was found to be closely associated with the malignant phenotype of hepatocellular carcinoma. However, its relationship with other malignancies needs to be further explored. Herein, we demonstrated that DNAJC24 exhibited upregulated expression in LUAD tissue samples and predicted poor survival in LUAD patients. The upregulation of DNAJC24 expression promoted proliferation and invasion of LUAD cells in A549 and NCI-H1299 cell lines. Further studies revealed that DNAJC24 could regulate the PI3K/AKT signaling pathway by affecting AKT phosphorylation. In addition, a series of experiments such as Co-IP and mass spectrometry confirmed that DNAJC24 could directly interact with PCNA and promoted the malignant phenotypic transformation of LUAD. In conclusion, our results suggested that DNAJC24 played an important role in the progression of LUAD and may serve as a specific prognostic biomarker for LUAD patients. The DNAJC24/PCNA/AKT axis may be a potential target for future individualized and precise treatment of LUAD patients.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ran Zuo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wei Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuchao He
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yu Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ping Yue
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenchen Gong
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jinfang Cui
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fuyi Zhu
- Department of Oncology Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yi Luo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lisha Qi
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yan Guo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Cancer Biobank of Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Liwei Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guangtao Li
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhiyong Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Peng Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Guo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
46
|
Du X, Cheng C, Yang Y, Fan B, Wang P, Xia H, Ni X, Liu Q, Lu L, Wei L. NSUN2 promotes lung adenocarcinoma progression through stabilizing PIK3R2 mRNA in an m 5C-dependent manner. Mol Carcinog 2024; 63:962-976. [PMID: 38411298 DOI: 10.1002/mc.23701] [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: 11/02/2023] [Revised: 01/23/2024] [Accepted: 02/02/2024] [Indexed: 02/28/2024]
Abstract
It is well known that 5-methylcytosine (m5C) is involved in variety of crucial biological processes in cancers. However, its biological roles in lung adenocarcinoma (LAUD) remain to be determined. The LUAD samples were used to assess the clinical value of NOP2/Sun RNA Methyltransferase 2 (NSUN2). Dot blot was used to determine global m5C levels. ChIP and dual-luciferase assays were performed to investigate the MYC-associated zinc finger protein (MAZ)-binding sites in NSUN2 promoter. RNA-seq was used to explore the downstream molecular mechanisms of NSUN2. Dual luciferase reporter assay, m5C-RIP-qPCR, and mRNA stability assay were conducted to explore the effect of NSUN2-depletion on target genes. Cell viability, transwell, and xenograft mouse model were designed to demonstrate the characteristic of NSUN2 in promoting LUAD progression. The m5C methyltransferase NSUN2 was highly expressed and caused elevated m5C methylation in LUAD samples. Mechanistically, MAZ positively regulated the transcription of NSUN2 and was related to poor survival of LUAD patients. Silencing NSUN2 decreased the global m5C levels, suppressed proliferation, migration and invasion, and inhibited activation of PI3K-AKT signaling in A549 and SPAC-1 cells. Phosphoinositide-3-Kinase Regulatory Subunit 2 (PIK3R2) was upregulated by NSUN2-mediated m5C methylation by enhancing its mRNA stabilization and activated the phosphorylation of the PI3K-AKT signaling. The present study explored the underlying mechanism and biological function of NSUN2-meditated m5C RNA methylation in LUAD. NSUN2 was discovered to facilitate the malignancy progression of LUAD through regulating m5C modifications to stabilize PIK3R2 activating the PI3K-AKT signaling, suggesting that NSUN2 could be a novel biomarker and promising therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Xuan Du
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Cheng
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Yang
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bowen Fan
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peiwen Wang
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haibo Xia
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Qizhan Liu
- Jiangsu Key Lab of Cancer Biomarkers, School of Public Health, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Lu
- Animal Core facility, The Key Laboratory of Model Animal, Jiangsu Animal Experimental Center for Medical and Pharmaceutical Research, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Wei
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
47
|
XIONG B, KE W, JIANG W. [Advances of Molecular Targeted Therapy in EGFR-mutated Squamous Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:283-290. [PMID: 38769831 PMCID: PMC11110274 DOI: 10.3779/j.issn.1009-3419.2024.101.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Indexed: 05/22/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a prevalent tumour type in our country, with lung squamous carcinoma being a commonly observed NSCLC subtype besides lung adenocarcinoma. Epidermal growth factor receptor (EGFR) is a significant driver gene in lung cancer, and EGFR mutation frequency is considerably lower in lung squamous carcinoma in comparison to lung adenocarcinoma. Although targeted therapy against EGFR has demonstrated significant advancements in lung adenocarcinoma, while progress in lung squamous carcinoma has been relatively sluggish. This paper reviews recent studies on molecular targeted therapy for EGFR-mutated lung squamous carcinoma and summarises the efficacy of EGFR-tyrosine kinase inhibitors (TKIs) in treating squamous carcinoma of the lung, in order to provide a reference for treating patients with EGFR-mutated squamous carcinoma of the lung.
.
Collapse
|
48
|
Miao Y, Wu L, Qiang J, Qi J, Li Y, Li R, Kong X, Zhang Q. The application of Raman spectroscopy for the diagnosis and monitoring of lung tumors. Front Bioeng Biotechnol 2024; 12:1385552. [PMID: 38699434 PMCID: PMC11063270 DOI: 10.3389/fbioe.2024.1385552] [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: 02/13/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Raman spectroscopy is an optical technique that uses inelastic light scattering in response to vibrating molecules to produce chemical fingerprints of tissues, cells, and biofluids. Raman spectroscopy strategies produce high levels of chemical specificity without requiring extensive sample preparation, allowing for the use of advanced optical tools such as microscopes, fiber optics, and lasers that operate in the visible and near-infrared spectral range, making them increasingly suitable for a wide range of medical diagnostic applications. Metal nanoparticles and nonlinear optical effects can improve Raman signals, and optimized fiber optic Raman probes can make real-time, in vivo, single-point observations. Furthermore, diagnostic speed and spatial accuracy can be improved through the multimodal integration of Raman measurements and other technologies. Recent studies have significantly contributed to the improvement of diagnostic speed and accuracy, making them suitable for clinical application. Lung cancer is a prevalent type of respiratory malignancy. However, the use of computed tomography for detection and screening frequently reveals numerous smaller lung nodules, which makes the diagnostic process more challenging from a clinical perspective. While the majority of small nodules detected are benign, there are currently no direct methods for identifying which nodules represent very early-stage lung cancer. Positron emission tomography and other auxiliary diagnostic methods for non-surgical biopsy samples from these small nodules yield low detection rates, which might result in significant expenses and the possibility of complications for patients. While certain subsets of patients can undergo curative treatment, other individuals have a less favorable prognosis and need alternative therapeutic interventions. With the emergence of new methods for treating cancer, such as immunotherapies, which can potentially extend patient survival and even lead to a complete cure in certain instances, it is crucial to determine the most suitable biomarkers and metrics for assessing the effectiveness of these novel compounds. This will ensure that significant treatment outcomes are accurately measured. This review provides a comprehensive overview of the prospects of Raman spectroscopy and its applications in the diagnosis and analysis of lung tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
49
|
Khashei Varnamkhasti K, Moghanibashi M, Naeimi S. Implications of ZNF334 gene in lymph node metastasis of lung SCC: potential bypassing of cellular senescence. J Transl Med 2024; 22:372. [PMID: 38637790 PMCID: PMC11025273 DOI: 10.1186/s12967-024-05115-9] [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: 11/01/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND The primary goal of this work is to identify biomarkers associated with lung squamous cell carcinoma and assess their potential for early detection of lymph node metastasis. METHODS This study investigated gene expression in lymph node metastasis of lung squamous cell carcinoma using data from the Cancer Genome Atlas and R software. Protein-protein interaction networks, hub genes, and enriched pathways were analyzed. ZNF334 and TINAGL1, two less explored genes, were further examined through in vitro, ex vivo, and in vivo experiments to validate the findings from bioinformatics analyses. The role of ZNF334 and TINAGL1 in senescence induction was assessed after H2O2 and UV induced senescence phenotype determined using β-galactosidase activity and cell cycle status assay. RESULTS We identified a total of 611 up- and 339 down-regulated lung squamous cell carcinoma lymph node metastasis-associated genes (FDR < 0.05). Pathway enrichment analysis highlighted the central respiratory pathway within mitochondria for the subnet genes and the nuclear DNA-directed RNA polymerases for the hub genes. Significantly down regulation of ZNF334 gene was associated with malignancy lymph node progression and senescence induction has significantly altered ZNF334 expression (with consistency in bioinformatics, in vitro, ex vivo, and in vivo results). Deregulation of TINAGL1 expression with inconsistency in bioinformatics, in vitro (different types of lung squamous cancer cell lines), ex vivo, and in vivo results, was also associated with malignancy lymph node progression and altered in senescence phenotype. CONCLUSIONS ZNF334 is a highly generalizable gene to lymph node metastasis of lung squamous cell carcinoma and its expression alter certainly under senescence conditions.
Collapse
Affiliation(s)
| | - Mehdi Moghanibashi
- Department of Genetics, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran.
| | - Sirous Naeimi
- Department of Genetics, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| |
Collapse
|
50
|
Cao F, Li M, Wang W, Yi Y, Chen Y, Liu H. A coumarin-furoxan hybrid as novel nitric oxide donor induced cell apoptosis and ferroptosis in NSCLC by promoting S-nitrosylation of STAT3 and negative regulation of JAK2-STAT3 pathway. Biochem Pharmacol 2024; 222:116068. [PMID: 38387529 DOI: 10.1016/j.bcp.2024.116068] [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: 10/31/2023] [Revised: 12/20/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Non-small cell lung cancer (NSCLC) still lacks effective treatment because of its extensive mutation diversity and frequent drug resistance. Therefore, it is urgent to develop new therapeutic strategies for NSCLC. In this study, we evaluated the inhibitory effect of a new coumarin-furoxan hybrid compound 9, a nitric oxide (NO) donor drug, on NSCLC proliferation and its mechanism. Our results show that compound 9 can inhibit the growth of four NSCLC cell lines and H1975 xenograft model in a dose-dependent manner. Compound 9 effectively releases high concentrations of NO within the mitochondria, leading to cellular oxidative stress, mitochondrial dysfunction, and apoptosis. Moreover, compound 9 inhibits JAK2/STAT3 protein phosphorylation and induces S-nitrosylation modification of STAT3, ultimately resulting in endogenous apoptosis in NSCLC. Additionally, compound 9 significantly induces NSCLC ferroptosis by depleting intracellular GSH, elevating MDA levels, inhibiting SLC7A11/GSH protein expression, and negatively regulating the JAK2/STAT3 pathway. In summary, this study elucidates the inhibitory effects of compound 9 on NSCLC proliferation and provides insights into the underlying mechanisms, offering new possibilities for NSCLC treatment strategies.
Collapse
Affiliation(s)
- Fan Cao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mengru Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weijie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Yi
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ying Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|