1
|
Xi S, Shan J, Wu X, Wang H, Zhang MR, Oyetunji S, Xu H, Xiao Z, Tolunay T, Carr SR, Hoang CD, Schrump DS. Repression of ZNFX1 by LncRNA ZFAS1 mediates tobacco-induced pulmonary carcinogenesis. Cell Mol Biol Lett 2025; 30:44. [PMID: 40211119 PMCID: PMC11983736 DOI: 10.1186/s11658-025-00705-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: 08/27/2024] [Accepted: 02/18/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Despite exhaustive research efforts, integrated genetic and epigenetic mechanisms contributing to tobacco-induced initiation and progression of lung cancers have yet to be fully elucidated. In particular, limited information is available regarding dysregulation of noncoding RNAs during pulmonary carcinogenesis. METHODS We examined correlations and interactions of long noncoding (lnc) RNAs and protein-coding genes in normal respiratory epithelial cells (NREC) and pulmonary tumor cells following exposure to cigarette smoke condensate (CSC) using gene expression arrays, qRT-PCR, western blot, growth assays, transwell assays, and murine xenograft models, as well as methylated DNA immunoprecipitation, RNA cross-link immunoprecipitation, and quantitative chromatin immunoprecipitation techniques with bioinformatics analyses. RESULTS Among diverse alterations of lncRNA and coding gene expression profiles in NREC exposed to CSC, we observed upregulation of lncRNA ZFAS1 and repression of an adjacent protein-coding gene, ZNFX1, and confirmed these findings in primary lung cancers. Phenotypic experiments indicated that ZFAS1 is an oncogene, whereas ZNFX1 functions as a tumor suppressor in lung cancer cells. Mechanistically, CSC induces ZFAS1 expression via SP1 and NFĸB-associated activation of an enhancer linked to ZFAS1. Subsequently, ZFAS1 interacts with DNA methyltransferases and polycomb group proteins to silence ZNFX1. Mithramycin and methysticin repress ZFAS1 and upregulate ZNFX1 in lung cancer cells in vitro and in vivo. CONCLUSION These studies reveal a novel feedforward lncRNA circuit contributing to pulmonary carcinogenesis and suggest that pharmacologic targeting of SP1 and/or NFĸB may be useful strategies for restoring ZNFX1 expression for lung tumor therapy.
Collapse
Affiliation(s)
- Sichuan Xi
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jigui Shan
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Xinwei Wu
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Haitao Wang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Mary R Zhang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Shakirat Oyetunji
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Hong Xu
- Laboratory of Cancer Prevention, National Cancer Institute, Frederick, MD, 21702, USA
| | - Zuoxiang Xiao
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Tuana Tolunay
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Shamus R Carr
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Chuong D Hoang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA
| | - David S Schrump
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Building 10; 4-3942, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Cangut B, Akinlusi R, Mohseny A, Ghesani N, Ghesani M. Evolving Paradigms in Lung Cancer: Latest Trends in Diagnosis, Management, and Radiopharmaceuticals. Semin Nucl Med 2025; 55:264-276. [PMID: 40055050 DOI: 10.1053/j.semnuclmed.2025.02.005] [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: 01/15/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025]
Abstract
Lung cancer is one of the most common and deadliest forms of cancer worldwide. Over the past two decades, significant changes have occurred in the classification of lung cancer, involving multidisciplinary input and emphasizing the growing contribution of immunohistochemistry and molecular techniques to morphology in the classification scheme. This comprehensive review will cover the background and epidemiology of lung cancer as well as advancements in its staging and management, including discussions of new surgical techniques, targeted therapies, and immunotherapy. The review will detail the role of 18F-FDG-PET-CT in lung cancer, highlighting its importance in staging, treatment response assessment, and recurrence detection. While immunotherapy has transformed lung cancer management and improved patient outcomes, it presents major challenges and opportunities for optimal assessment of treatment response in lung cancer patients using 18F-FDG-PET-CT. This review will also explore future directions, including a discussion of promising new targeted diagnostic radiopharmaceuticals for PET/CT imaging. Additionally, there will be a brief discussion of evolving and exciting treatment options for lung cancer using targeted therapeutic radiopharmaceuticals. Several case-based illustrations are included to exemplify the role of 18F-FDG-PET-CT in various clinical scenarios.
Collapse
Affiliation(s)
- Busra Cangut
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rahman Akinlusi
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ava Mohseny
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nasrin Ghesani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Munir Ghesani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
3
|
Narra K, Ghabach B, Athipatla V, Blackwell JM, Teigen KJ, Bullock JC, Diaz A, Gerber DE, von Itzstein MS. Identification and Treatment of Lung Cancer Oncogenic Drivers in a Diverse Safety Net Setting. Clin Lung Cancer 2025; 26:83-92. [PMID: 39304363 DOI: 10.1016/j.cllc.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/07/2024] [Accepted: 08/24/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Advances in the testing and treatment of patients with non-small cell lung cancer (NSCLC) harboring oncogenic drivers have improved outcomes. Little is known about testing and treatment patterns in diverse patient populations. METHODS We conducted a retrospective study in a diverse cohort of patients treated in the John Peter Smith safety net healthcare system. We determined patterns of blood- and tissue-based testing and treatment of patients with EGFR and ALK alterations. Cox proportional-hazards regression models were used to assess the impact of EGFR and ALK testing. RESULTS A total of 220 patients were included, 97 (44%) were non-Hispanic White, 72 (33%) were Black, 28 (13%) were Hispanic, and 23 (10%) were Asian. EGFR and ALK testing increased over time from 55% and 52%, respectively, in 2017 to 87% and 82%, respectively, in 2021. Frequency of EGFR alterations were highest in Asian patients (45%) and comparable among other groups (6-13%). Frequency of ALK alterations were highest in Hispanic (13%), and Asian (11%) patients, and were 2% for both Black and non-Hispanic White patients. In a multivariate model, lack of testing was associated with worse survival (aHR 1.6; P = .003) and testing positive for EGFR (aHR 0.43; P = .01) or ALK (aHR 0.28; P = .04) was associated with improved survival. Race and ethnicity were not associated with survival differences. CONCLUSION As molecular testing for oncogenic mutations in NSCLC increases, druggable alterations such as ALK and EGFR can be identified in all race-ethnicity groups and are associated with improved outcomes.
Collapse
Affiliation(s)
- Kalyani Narra
- John Peter Smith Oncology and Infusion Center, Fort Worth, TX; Department of Internal Medicine, Burnett School of Medicine at Texas Christian University, Fort Worth, TX.
| | - Bassam Ghabach
- John Peter Smith Oncology and Infusion Center, Fort Worth, TX; Department of Internal Medicine, Burnett School of Medicine at Texas Christian University, Fort Worth, TX
| | | | | | - Kari J Teigen
- Office of Clinical Research, John Peter Smith Health Network, Fort Worth, TX
| | | | - Anna Diaz
- Office of Clinical Research, John Peter Smith Health Network, Fort Worth, TX
| | - David E Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX; Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX; Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mitchell S von Itzstein
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX; Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
4
|
Garitaonaindia Y, Martínez-Cutillas M, Uribarren M, Redondo I, Calvo V, Serna-Blasco R, Provencio M. Adoptive cell therapies in thoracic malignancies: a comprehensive review. Clin Transl Oncol 2025:10.1007/s12094-024-03834-5. [PMID: 39789380 DOI: 10.1007/s12094-024-03834-5] [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: 09/20/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
This review aims to summarize recent studies and findings within adoptive cell therapies, including tumor-infiltrating lymphocytes, genetically engineered T cell receptors, and chimeric antigen receptor T cells, in the treatment of thoracic malignancies, including non-small cell lung cancer, small cell lung cancer, and malignant pleural mesothelioma. Several trials are ongoing, and a few have reported results, suggesting that adoptive cell therapies may represent a potential treatment option for these patients, especially when checkpoint inhibition has failed. We also discuss the potential implementation of these therapies, as they present a new toxicity profile and an intrinsic financial burden. Despite the challenges to overcome, such as the accurate identification of antigens and developing strategies to improve efficacy and toxicity profiles, new cellular therapies are experiencing significant development in the field of thoracic malignancies.
Collapse
Affiliation(s)
- Yago Garitaonaindia
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain.
| | - Marta Martínez-Cutillas
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Maria Uribarren
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Isabel Redondo
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Virginia Calvo
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Roberto Serna-Blasco
- Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Puerta De Hierro University Hospital, Majadahonda, Madrid, Spain
| | - Mariano Provencio
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain.
| |
Collapse
|
5
|
Chayangsu C, Khorana J, Charoentum C, Sriuranpong V, Patumanond J, Tantraworasin A. Exploring Prognostic Factors and Survival Outcomes in Advanced Non-Small Cell Lung Cancer Patients Undergoing First-Line Chemotherapy in Limited-Resource Settings. J Clin Med 2025; 14:335. [PMID: 39860343 PMCID: PMC11766160 DOI: 10.3390/jcm14020335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality globally, especially in limited-resource countries (LRCs) where access to advanced treatments such as targeted therapy and immunotherapy is constrained. Platinum-based chemotherapy remains a cornerstone of first-line therapy. This study aims to identify prognostic factors influencing survival outcomes and evaluate treatment response to chemotherapy in advanced NSCLC patients in LRCs. Methods: A retrospective cohort study was conducted on 200 advanced NSCLC patients treated with first-line platinum-based doublet chemotherapy at Surin Hospital Cancer Center, Thailand. Prognostic factors were assessed through univariate and multivariate Cox regression analyses. Additionally, restricted mean survival time (RMST) was calculated to compare survival outcomes between responders and non-responders. Results: Independent prognostic factors associated with improved survival included good performance status, ECOG 0-1 (HR 0.50, p = 0.012), serum albumin ≥ 3.5 mg/dL (HR 0.60, p = 0.010), and favorable response to chemotherapy (HR 0.57, p = 0.003). Responders demonstrated significantly longer RMST at 12 months (p < 0.001), 24 months (p < 0.001), and 36 months (p = 0.004) compared to non-responders. Conclusions: Identifying prognostic factors and treatment responses is important for improving outcomes in advanced NSCLC patients, particularly in limited-resource settings where access to novel therapies is restricted.
Collapse
Affiliation(s)
- Chawalit Chayangsu
- Department of Internal Medicine, Surin Hospital, Institute of Medicine, Suranaree University of Technology, Surin 32000, Thailand;
| | - Jiraporn Khorana
- Department of Surgery, Division of Pediatric Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Clinical Epidemiology and Clinical Statistic Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Chaiyut Charoentum
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Virote Sriuranpong
- Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Jayanton Patumanond
- Clinical Epidemiology and Clinical Statistic Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Apichat Tantraworasin
- Clinical Epidemiology and Clinical Statistic Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Clinical Surgical Research Center, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
6
|
Peng L, Xia L, Yang M, Wen Y, Zeng Q. Comprehensive bioinformatics analysis of prognosis and immunotherapy in lung adenocarcinoma. J Thorac Dis 2024; 16:8633-8647. [PMID: 39831256 PMCID: PMC11740037 DOI: 10.21037/jtd-24-1530] [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: 09/14/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
Background Research has shown that genetic mutations play an important role in the prognosis of lung adenocarcinoma (LUAD). However, the genes that influence the prognosis and immunotherapy of lung cancer patients have not yet been thoroughly studied. In this study, data from The Cancer Genome Atlas (TCGA) Program and other databases were used to identify the survival-related genes in LUAD. Methods First, the TCGA database was used to screen key LUAD genes. Second, the Gene Expression Profiling Interactive Analysis 2 (GEPIA2), University of ALabama at Birmingham CANcer (UALCAN), Tumor IMmune Estimation Resource (TIMER), Kaplan-Meier plotter, and cBioPortal databases, and a univariate Cox analysis combined with a random forest (RF) model were used to estimate gene expression, patient prognosis, and gene mutations, respectively. TIMER was also used to predict the immune function of the genes. Results A total of 2,138 up-regulated and 2,559 down-regulated differentially expressed genes (DEGs) were identified from TCGA-LUAD dataset. Next, four prognostic genes (i.e., CENPH, SLC35F4, TESMIN, and TERT) were identified as the key genes. The expression levels of all four genes were higher in LUAD tissues than those in the normal lung tissues, but only CENPH and TESMIN were correlated with poor overall survival (OS). The four genes were also found to be associated with immunoinfiltration. Conclusions Of the four key genes identified, CENPH and TESMIN would not only contribute to the diagnosis and prognosis of LUAD but could also serve as potential immunotherapy targets for LUAD.
Collapse
Affiliation(s)
- Ling Peng
- Department of Respiratory and Critical Care Medicine, Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Luping Xia
- China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang, China
- Department of Respiratory and Critical Care Medicine, Shangrao People’s Hospital, Shangrao, China
| | - Meiyu Yang
- Department of Respiratory and Critical Care Medicine, Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yali Wen
- Department of Respiratory and Critical Care Medicine, Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qinghua Zeng
- Department of Respiratory and Critical Care Medicine, Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Qian L, Ji Z, Mei L, Zhao J. IGF2BP2 promotes lung adenocarcinoma progression by regulating LOX1 and tumor-associated neutrophils. Immunol Res 2024; 73:16. [PMID: 39688738 DOI: 10.1007/s12026-024-09563-9] [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: 09/21/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024]
Abstract
Lung adenocarcinoma (LUAD) is the common form of lung cancer and is prone to distant metastasis. IGF2BP2, an m6A modification regulator, is upregulated in lung cancer tissue, but its specific role within the LUAD tumor microenvironment (TME) is unknown. We explored the role of IGF2BP2 in the progression of LUAD. IGF2BP2 expression in LUAD patient specimens and controls was evaluated through bioinformatics, Western blot, and immunohistochemistry. LUAD subcutaneous and orthotopic xenograft tumor models were established, alongside a co-culture system of tumor-associated neutrophils (TANs) and A549 cells. Functional assays assessed IGF2BP2's role under treatment with JX5, an IGF2BP2 inhibitor. Mechanistic assays explored the interaction between IGF2BP2 and LOX1 in 293T cells. IGF2BP2 was significantly upregulated in LUAD tissues, with higher expression levels predicting worse prognosis for patients (p < 0.001). In subcutaneous and orthotopic xenograft models, treatment with JX5, an IGF2BP2 inhibitor, reduced tumor size, volume, and weight (p < 0.001). JX5 also significantly reduced the concentrations of pro-inflammatory cytokines in peripheral blood (p < 0.01 and p < 0.001). Flow cytometry analysis indicated JX5 reduced CD11b+Ly6G+/CD45+ cells (TANs) in the TME (p < 0.001). Mechanistically, JX5 downregulated LOX1 expression in vivo, and co-culture experiments further demonstrated that IGF2BP2 promotes LUAD progression through LOX1-mediated regulation of TAN activity (p < 0.01 and p < 0.001). Overexpression of LOX1 reversed the inhibitory effects of JX5 on TAN infiltration, tumor cell viability, and apoptosis (p < 0.01 and p < 0.001). Additionally, RNA immunoprecipitation revealed that IGF2BP2 binds to LOX1 mRNA at its m6A modification site, stabilizing LOX1 and enhancing its function in the TME. Knockdown of IGF2BP2 accelerated LOX1 mRNA degradation, confirming its role in maintaining LOX1 stability (p < 0.01 and p < 0.001). IGF2BP2 recognizes and stabilizes LOX1 through m6A modification, contributing to TAN-mediated LUAD progression. Overall, these findings offer new insights into LUAD progression and demonstrate that IGF2BP2 is a key regulator that promotes tumor advancement, highlighting the IGF2BP2-LOX1 axis as a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Long Qian
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu, China
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Zhuqing Ji
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Medical Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Lingyun Mei
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
8
|
Helmy MW, Youssef MH, Yamari I, Amr A, Moussa FI, El Wakil A, Chtita S, El-Samad LM, Hassan MA. Repurposing of sericin combined with dactolisib or vitamin D to combat non-small lung cancer cells through computational and biological investigations. Sci Rep 2024; 14:27034. [PMID: 39505930 PMCID: PMC11541877 DOI: 10.1038/s41598-024-76947-0] [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: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
This study aims to repurpose sericin in combating non-small lung cancer cells (A549 and H460) by combining it with dactolisib or vitamin D to reduce the dose of dactolisib and boost the anticancer effectiveness of dactolisib and vitamin D. Therefore, the binding affinities of individual and combined drugs were examined using in silico and protein-protein interaction studies, targeting NF-κB, Cyclin D1, p-AKT, and VEGF1 proteins. The findings manifested remarkable affinities for combinatorial drugs compared to individual compounds. To substantiate these findings, the combined IC50 for each combination (sericin + dactolisib and sericin + vitamin D) were determined, reporting 31.9 and 41.8 µg/ml, respectively, against A549 cells and 47.9 and 55.3 µg/ml, respectively, against H460 cells. Furthermore, combination indices were assessed to lower the doses of each drug. Interestingly, in vitro results exhibited marked diminutions in NF-κB, Cyclin D1, p-AKT, and VEGF1 after treatment with sericin + dactolisib and sericin + vitamin D compared to control lung cancer cells and those treated with a single drug. Moreover, A549 and H460 cells treated with both combinations demonstrated augmented caspase-3 levels, implying substantial apoptotic activity. Altogether, these results accentuated the prospective implementation of sericin in combination with dactolisib and vitamin D at low doses to preclude lung cancer cell proliferation.
Collapse
Affiliation(s)
- Maged W Helmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Damanhour, Egypt
| | - Mariam H Youssef
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Imane Yamari
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, P. O. Box 7955, Casablanca, Morocco
| | - Alaa Amr
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Farouzia I Moussa
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Abeer El Wakil
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Alexandria, Egypt
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, P. O. Box 7955, Casablanca, Morocco
| | - Lamia M El-Samad
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Mohamed A Hassan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt.
| |
Collapse
|
9
|
Ntzifa A, Marras T, Kallergi G, Kotsakis A, Georgoulias V, Lianidou E. Comprehensive liquid biopsy analysis for monitoring NSCLC patients under second-line osimertinib treatment. Front Oncol 2024; 14:1435537. [PMID: 39497713 PMCID: PMC11532185 DOI: 10.3389/fonc.2024.1435537] [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: 05/20/2024] [Accepted: 09/09/2024] [Indexed: 11/07/2024] Open
Abstract
Background The heterogeneous and complex genetic landscape of NSCLC impacts the clinical outcomes of patients who will eventually develop resistance to osimertinib. Liquid biopsy (LB) analysis as a minimally invasive approach is a key step to efficiently identify resistance mechanisms and adjust to proper subsequent treatments. Materials and methods In the present study, we combined plasma-cfDNA and CTC analysis from 30 NSCLC patients in samples collected before treatment and at the progression of disease (PD). We detected molecular alterations at the DNA mutation (EGFR, PIK3CA, KRAS G12C, BRAF V600E), DNA methylation (RASSF1A, BRMS1, FOXA1, SLFN1, SHISA3, RARβ,, WIF-1, RASSF10 and APC), gene expression (CK-19, CK-18, CK-8, AXL, TWIST-1, PD-L1, PIM-1, Vimentin, ALDH-1, and B2M) and chromosomal level (HER2 and MET amplification) as possible resistance mechanisms and druggable targets. We also studied the expression of PD-L1 in single CTCs using immunofluorescence. Results In some cases, T790M resistance EGFR mutation was detected at baseline in CTCs but not in the corresponding plasma cfDNA. PIK3CA mutations were detected only in plasma-cfDNA but not in corresponding CTCs. KRAS G12C and BRAF V600E mutations were not detected in the samples analyzed. MET amplification was detected in the CTCs of two patients before treatment whereas HER2 amplification was detected in the CTCs of three patients at baseline and in one patient at PD. DNA methylation analysis revealed low concordance between CTCs and cfDNA, indicating the complementary information obtained through parallel LB analysis. Results from gene expression analysis indicated high rates of vimentin-positive CTCs detected at all time points during osimertinib. Moreover, there was an increased number of NSCLC patients at PD harboring CTCs positive in PD-L1. AXL and PIM-1 expression detected in CTCs during treatment suggesting new possible therapeutic strategies. Discussion Our results reveal that comprehensive liquid biopsy analysis can efficiently represent the heterogeneous molecular landscape and provide prominent information on subsequent treatments for NSCLC patients at PD since druggable molecular alterations were detected in CTCs.
Collapse
Affiliation(s)
- Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Marras
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Athanasios Kotsakis
- Department of Medical Oncology, General University Hospital of Larissa, Larissa, Greece
| | - Vasilis Georgoulias
- First Department of Medical Oncology, Metropolitan General Hospital of Athens, Cholargos, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Danbala IA, Fu S, Sheng W, Tang H, Magashi MA, Wang X. Immune checkpoint inhibitors with or without radiotherapy in metastatic non‑small cell lung cancer: A meta‑analysis and literature review. Oncol Lett 2024; 28:489. [PMID: 39185490 PMCID: PMC11342421 DOI: 10.3892/ol.2024.14622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 07/03/2024] [Indexed: 08/27/2024] Open
Abstract
The combination of immune checkpoint inhibitors (ICIs) and radiotherapy has shown promise in the treatment of metastatic non-small cell lung cancer (NSCLC). The present meta-analysis aimed to determine the efficacy and safety of combining radiotherapy (RT) ICIs for the treatment of metastatic NSCLC. PubMed, Google Scholar, the Cochrane Library and Web of Science databases were searched for relevant articles up to February 1, 2023. Post-therapy outcomes such as progression-free survival (PFS), complete response, partial response (PR), progressive disease (PD), stable disease and adverse events (AEs) were analyzed. The meta-analysis was performed using RevMan 5.4 software. A total of seven studies involving 682 patients were included (384 patients who received ICI + RT vs. RT and 298 patients who received ICI + RT vs. ICI alone). No significant difference in PFS was demonstrated between the ICI + RT group and the RT group (heterogeneity: χ2=2.35; df=1; P=0.13; I2=57% and test for overall effect: Z=0.10; P=0.92). Conversely, patients in the ICI alone group had significantly decreased PR rates (heterogeneity: Τ2=0.00; χ2=2.13; df=3; P=0.54; I2=0% and test for overall effect: Z=2.57; P=0.01) compared with patients in the ICI + RT group. The ICI + RT group also had significantly lower rates of PD (heterogeneity: Τ2=0.00; χ2=0.91; df=3; P=0.82; I2=0% and test for overall effect: Z=2.52; P=0.01) compared with the ICI alone group. Safety analysis revealed no significant difference between patients who received ICI + RT and those who received RT in terms of grade 1 or 2 AEs. In conclusion, the combination of ICIs + RT demonstrates promising efficacy and safety for patients with metastatic NSCLC. However, clinical trials that have tested this combination are lacking, which emphasizes the need for further research.
Collapse
Affiliation(s)
- Isah Adamu Danbala
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
- Overseas Education College, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Shengqiao Fu
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Wanying Sheng
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Haowen Tang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Mahmud Abdulkadir Magashi
- Overseas Education College, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
- Department of Gastrointestinal Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Xu Wang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
11
|
Miranda EI, Gierbolini-Bermúdez A, Quintana R, Torres-Cintrón CR, Ortiz-Ortiz KJ. Treatment Patterns and Health Care Resource Utilization of Patients With Non-Small Cell Lung Cancer in Puerto Rico: The TREATLINES-ONCOLUNG Study. JCO Glob Oncol 2024; 10:e2400089. [PMID: 39348632 DOI: 10.1200/go.24.00089] [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: 02/26/2024] [Revised: 07/10/2024] [Accepted: 08/07/2024] [Indexed: 10/02/2024] Open
Abstract
PURPOSE Lung cancer remains one of the leading causes of cancer-related mortality worldwide. It is the third cause of death among patients with cancer in Puerto Rico (PR) and non-small cell lung cancer (NSCLC) is the most prevalent. This study aims to describe the first-line treatment (1LT) and health care resource utilization (HCRU) among patients with NSCLC in PR. METHODS A retrospective cohort study was conducted using the PR Central Cancer Registry Health Insurance Linkage Database to describe patients with NSCLC from 2012 to 2016. It describes sociodemographic and clinical characteristics on the basis of stage and histology and includes 1LT patterns and HCRU. RESULTS A total of 1,011 patients met the inclusion criteria. Most were male (57.1%), married (54.1%), and had no comorbidities (55.8%). A significant proportion of patients (71.1%) were diagnosed at stages III and IV, with nonsquamous cell carcinoma being the most prevalent histology group (75.9%). About 61.7% received systemic therapy, 36.7% received radiotherapy, and 21.9% underwent surgery. Platinum (Pt)-based combinations were the most common 1LT (82.9%). On average, patients had 4.7 emergency room visits, nearly six hospitalizations, and 22.4 outpatient visits annually. The mean frequencies of positron emission tomography, ultrasounds, computerized tomography scans, and magnetic resonance imaging were 0.95, 0.11, 4.88, and 0.91, respectively. CONCLUSION To our knowledge, this study provides the first description of 1LT patterns, HCRU, and sociodemographic information among patients with NSCLC in PR. A significant number of patients were diagnosed at stage III or higher and received Pt-based systemic therapy as their 1LT. More research is required to investigate treatment patterns beyond the 1LT and to gain a comprehensive understanding of optimal care interventions and factors associated with early NSCLC diagnosis.
Collapse
Affiliation(s)
| | - Axel Gierbolini-Bermúdez
- Division of Cancer Control and Population Sciences, Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Puerto Rico Central Cancer Registry, Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
| | | | - Carlos R Torres-Cintrón
- Puerto Rico Central Cancer Registry, Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
| | - Karen J Ortiz-Ortiz
- Division of Cancer Control and Population Sciences, Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Puerto Rico Central Cancer Registry, Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Health Services Administration, Graduate School of Public Health, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
12
|
Ntzifa A, Marras T, Georgoulias V, Lianidou E. Liquid biopsy for the management of NSCLC patients under osimertinib treatment. Crit Rev Clin Lab Sci 2024; 61:347-369. [PMID: 38305080 DOI: 10.1080/10408363.2024.2302116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
Therapeutic management of NSCLC patients is quite challenging as they are mainly diagnosed at a late stage of disease, and they present a high heterogeneous molecular profile. Osimertinib changed the paradigm shift in treatment of EGFR mutant NSCLC patients achieving significantly better clinical outcomes. To date, osimertinib is successfully administered not only as first- or second-line treatment, but also as adjuvant treatment while its efficacy is currently investigated during neoadjuvant treatment or in stage III, unresectable EGFR mutant NSCLC patients. However, resistance to osimertinib may occur due to clonal evolution, under the pressure of the targeted therapy. The utilization of liquid biopsy as a minimally invasive tool provides insight into molecular heterogeneity of tumor clonal evolution and potent resistance mechanisms which may help to develop more suitable therapeutic approaches. Longitudinal monitoring of NSCLC patients through ctDNA or CTC analysis could reveal valuable information about clinical outcomes during osimertinib treatment. Therefore, several guidelines suggest that liquid biopsy in addition to tissue biopsy should be considered as a standard of care in the advanced NSCLC setting. This practice could significantly increase the number of NSCLC patients that will eventually benefit from targeted therapies, such as EGFR TKIs.
Collapse
Affiliation(s)
- Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Marras
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasilis Georgoulias
- First Department of Medical Oncology, Metropolitan General Hospital of Athens, Cholargos, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
Shukla V, Wang H, Varticovski L, Baek S, Wang R, Wu X, Echtenkamp F, Villa-Hernandez F, Prothro KP, Gara SK, Zhang MR, Shiffka S, Raziuddin R, Neckers LM, Linehan WM, Chen H, Hager GL, Schrump DS. Genome-Wide Analysis Identifies Nuclear Factor 1C as a Novel Transcription Factor and Potential Therapeutic Target in SCLC. J Thorac Oncol 2024; 19:1201-1217. [PMID: 38583771 DOI: 10.1016/j.jtho.2024.03.023] [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: 03/14/2024] [Accepted: 03/30/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Recent insights regarding mechanisms mediating stemness, heterogeneity, and metastatic potential of lung cancers have yet to be fully translated to effective regimens for the treatment of these malignancies. This study sought to identify novel targets for lung cancer therapy. METHODS Transcriptomes and DNA methylomes of 14 SCLC and 10 NSCLC lines were compared with normal human small airway epithelial cells (SAECs) and induced pluripotent stem cell (iPSC) clones derived from SAEC. SCLC lines, lung iPSC (Lu-iPSC), and SAEC were further evaluated by DNase I hypersensitive site sequencing (DHS-seq). Changes in chromatin accessibility and depths of transcription factor (TF) footprints were quantified using Bivariate analysis of Genomic Footprint. Standard techniques were used to evaluate growth, tumorigenicity, and changes in transcriptomes and glucose metabolism of SCLC cells after NFIC knockdown and to evaluate NFIC expression in SCLC cells after exposure to BET inhibitors. RESULTS Considerable commonality of transcriptomes and DNA methylomes was observed between Lu-iPSC and SCLC; however, this analysis was uninformative regarding pathways unique to lung cancer. Linking results of DHS-seq to RNA sequencing enabled identification of networks not previously associated with SCLC. When combined with footprint depth, NFIC, a transcription factor not previously associated with SCLC, had the highest score of occupancy at open chromatin sites. Knockdown of NFIC impaired glucose metabolism, decreased stemness, and inhibited growth of SCLC cells in vitro and in vivo. ChIP-seq analysis identified numerous sites occupied by BRD4 in the NFIC promoter region. Knockdown of BRD4 or treatment with Bromodomain and extra-terminal domain (BET) inhibitors (BETis) markedly reduced NFIC expression in SCLC cells and SCLC PDX models. Approximately 8% of genes down-regulated by BETi treatment were repressed by NFIC knockdown in SCLC, whereas 34% of genes repressed after NFIC knockdown were also down-regulated in SCLC cells after BETi treatment. CONCLUSIONS NFIC is a key TF and possible mediator of transcriptional regulation by BET family proteins in SCLC. Our findings highlight the potential of genome-wide chromatin accessibility analysis for elucidating mechanisms of pulmonary carcinogenesis and identifying novel targets for lung cancer therapy.
Collapse
Affiliation(s)
- Vivek Shukla
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Present Address: Division of Nonclinical Sciences (DNCS), FDA, Silver Spring, Maryland
| | - Haitao Wang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lyuba Varticovski
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ruihong Wang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Xinwei Wu
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank Echtenkamp
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank Villa-Hernandez
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Katherine P Prothro
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sudheer K Gara
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary R Zhang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie Shiffka
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Razi Raziuddin
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Leonard M Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Haobin Chen
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Present Address: Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David S Schrump
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
14
|
Garg P, Singhal S, Kulkarni P, Horne D, Malhotra J, Salgia R, Singhal SS. Advances in Non-Small Cell Lung Cancer: Current Insights and Future Directions. J Clin Med 2024; 13:4189. [PMID: 39064229 PMCID: PMC11278207 DOI: 10.3390/jcm13144189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The leading cause of cancer deaths worldwide is attributed to non-small cell lung cancer (NSCLC), necessitating a continual focus on improving the diagnosis and treatment of this disease. In this review, the latest breakthroughs and emerging trends in managing NSCLC are highlighted. Major advancements in diagnostic methods, including better imaging technologies and the utilization of molecular biomarkers, are discussed. These advancements have greatly enhanced early detection and personalized treatment plans. Significant improvements in patient outcomes have been achieved by new targeted therapies and immunotherapies, providing new hope for individuals with advanced NSCLC. This review discusses the persistent challenges in accessing advanced treatments and their associated costs despite recent progress. Promising research into new therapies, such as CAR-T cell therapy and oncolytic viruses, which could further revolutionize NSCLC treatment, is also highlighted. This review aims to inform and inspire continued efforts to improve outcomes for NSCLC patients globally, by offering a comprehensive overview of the current state of NSCLC treatment and future possibilities.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Sulabh Singhal
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Departments of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Jyoti Malhotra
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
15
|
Horibe R, Yokota M, Uemura K, Hashimoto M, Kawagishi N, Nishiyama K. De novo Hepatitis B Virus Reactivation during Treatment with an Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor in a Patient with Advanced Lung Cancer. Intern Med 2024; 63:1797-1800. [PMID: 37926534 PMCID: PMC11239268 DOI: 10.2169/internalmedicine.2731-23] [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: 08/07/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
A 71-year-old woman was treated with osimertinib for stage IV adenocarcinoma with epidermal growth factor receptor (EGFR) mutations. Treatment led to improvements in the primary tumor, multiple lung metastases, and multiple bone metastases. However, nine months later, she presented with marked liver dysfunction and jaundice. Chest and abdominal computed tomography did not show abnormal findings in the liver parenchyma or biliary system. However, blood tests were positive for hepatitis B surface antigen and hepatitis B virus DNA, suggesting hepatitis B virus reactivation. The patient died of liver failure despite treatment with steroids and antiviral drugs.
Collapse
Affiliation(s)
- Ryota Horibe
- Department of Respiratory Medicine, NTT-East Corporation Sapporo Medical Center, Japan
| | - Motohiro Yokota
- Department of Respiratory Medicine, NTT-East Corporation Sapporo Medical Center, Japan
| | - Koujiro Uemura
- Department of Respiratory Medicine, NTT-East Corporation Sapporo Medical Center, Japan
| | - Midori Hashimoto
- Department of Respiratory Medicine, NTT-East Corporation Sapporo Medical Center, Japan
| | - Naoki Kawagishi
- Department of Gastroenterology, NTT-East Corporation Sapporo Medical Center, Japan
| | - Kaoru Nishiyama
- Department of Respiratory Medicine, NTT-East Corporation Sapporo Medical Center, Japan
| |
Collapse
|
16
|
Baniasadi A, Das JP, Prendergast CM, Beizavi Z, Ma HY, Jaber MY, Capaccione KM. Imaging at the nexus: how state of the art imaging techniques can enhance our understanding of cancer and fibrosis. J Transl Med 2024; 22:567. [PMID: 38872212 PMCID: PMC11177383 DOI: 10.1186/s12967-024-05379-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Both cancer and fibrosis are diseases involving dysregulation of cell signaling pathways resulting in an altered cellular microenvironment which ultimately leads to progression of the condition. The two disease entities share common molecular pathophysiology and recent research has illuminated the how each promotes the other. Multiple imaging techniques have been developed to aid in the early and accurate diagnosis of each disease, and given the commonalities between the pathophysiology of the conditions, advances in imaging one disease have opened new avenues to study the other. Here, we detail the most up-to-date advances in imaging techniques for each disease and how they have crossed over to improve detection and monitoring of the other. We explore techniques in positron emission tomography (PET), magnetic resonance imaging (MRI), second generation harmonic Imaging (SGHI), ultrasound (US), radiomics, and artificial intelligence (AI). A new diagnostic imaging tool in PET/computed tomography (CT) is the use of radiolabeled fibroblast activation protein inhibitor (FAPI). SGHI uses high-frequency sound waves to penetrate deeper into the tissue, providing a more detailed view of the tumor microenvironment. Artificial intelligence with the aid of advanced deep learning (DL) algorithms has been highly effective in training computer systems to diagnose and classify neoplastic lesions in multiple organs. Ultimately, advancing imaging techniques in cancer and fibrosis can lead to significantly more timely and accurate diagnoses of both diseases resulting in better patient outcomes.
Collapse
Affiliation(s)
- Alireza Baniasadi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA.
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Conor M Prendergast
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Zahra Beizavi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Hong Y Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | | | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| |
Collapse
|
17
|
Rosca OC, Vele OE. Microsatellite Instability, Mismatch Repair, and Tumor Mutation Burden in Lung Cancer. Surg Pathol Clin 2024; 17:295-305. [PMID: 38692812 DOI: 10.1016/j.path.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Since US Food and Drug Administration approval of programmed death ligand 1 (PD-L1) as the first companion diagnostic for immune checkpoint inhibitors (ICIs) in non-small cell lung cancer, many patients have experienced increased overall survival. To improve selection of ICI responders versus nonresponders, microsatellite instability/mismatch repair deficiency (MSI/MMR) and tumor mutation burden (TMB) came into play. Clinical data show PD-L1, MSI/MMR, and TMB are independent predictive immunotherapy biomarkers. Harmonization of testing methodologies, optimization of assay design, and results analysis are ongoing. Future algorithms to determine immunotherapy eligibility might involve complementary use of current and novel biomarkers. Artificial intelligence could facilitate algorithm implementation to convert complex genetic data into recommendations for specific ICIs.
Collapse
Affiliation(s)
- Oana C Rosca
- Molecular Pathologist/Cytopathologist, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Department of Pathology and Laboratory Medicine, 2200 Northern Boulevard, Suite 104, Greenvale, NY 11548, USA.
| | - Oana E Vele
- Molecular Pathologist/Cytopathologist, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Department of Pathology and Laboratory Medicine, Lenox Hill Hospital, New York, NY 10075, USA
| |
Collapse
|
18
|
Zhou D, Li Y, Liu Q, Deng X, Chen L, Li M, Zhang J, Lu X, Zheng H, Dai J. Integrated whole-exome and bulk transcriptome sequencing delineates the dynamic evolution from preneoplasia to invasive lung adenocarcinoma featured with ground-glass nodules. Cancer Med 2024; 13:e7383. [PMID: 38864483 PMCID: PMC11167609 DOI: 10.1002/cam4.7383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/15/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
OBJECTIVE The genomic and molecular ecology involved in the stepwise continuum progression of lung adenocarcinoma (LUAD) from adenocarcinoma in situ (AIS) to minimally invasive adenocarcinoma (MIA) and subsequent invasive adenocarcinoma (IAC) remains unclear and requires further elucidation. We aimed to characterize gene mutations and expression landscapes, and explore the association between differentially expressed genes (DEGs) and significantly mutated genes (SMGs) during the dynamic evolution from AIS to IAC. METHODS Thirty-five patients with ground-glass nodules (GGNs) lung adenocarcinomas were enrolled. Whole-exome sequencing (WES) and transcriptome sequencing (RNA-Seq) were conducted on all patients, encompassing both tumor samples and corresponding noncancerous tissues. Data obtained from WES and RNA-Seq were subsequently analyzed. RESULTS The findings from WES delineated that the predominant mutations were observed in EGFR (49%) and ANKRD36C (17%). SMGs, including EGFR and RBM10, were associated with the dynamic evolution from AIS to IAC. Meanwhile, DEGs, including GPR143, CCR9, ADAMTS16, and others were associated with the entire process of invasive LUAD. We found that the signaling pathways related to cell migration and invasion were upregulated, and the signaling pathways of angiogenesis were downregulated across the pathological stages. Furthermore, we found that the messenger RNA (mRNA) levels of FAM83A, MAL2, DEPTOR, and others were significantly correlated with CNVs. Gene set enrichment analysis (GSEA) showed that heme metabolism and cholesterol homeostasis pathways were significantly upregulated in patients with EGFR/RBM10 co-mutations, and these patients may have poorer overall survival than those with EGFR mutations. Based on the six calculation methods for the immune infiltration score, NK/CD8+ T cells decreased, and Treg/B cells increased with the progression of early LUAD. CONCLUSIONS Our findings offer valuable insights into the unique genomic and molecular features of LUAD, facilitating the identification and advancement of precision medicine strategies targeting the invasive progression of LUAD from AIS to IAC.
Collapse
Affiliation(s)
- Dong Zhou
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Yan‐qi Li
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Quan‐xing Liu
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Xu‐feng Deng
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Liang Chen
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Man‐yuan Li
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Jiao Zhang
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Xiao Lu
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Hong Zheng
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Ji‐gang Dai
- Department of Thoracic SurgeryXinqiao Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| |
Collapse
|
19
|
Ginés Rubió J, Delgado O, Callejo A, Domínguez M, Torres C. Healthcare Resource Utilization and Associated Costs among Patients with Advanced Non-Small-Cell Lung Cancer Receiving Chemotherapy or Immunotherapy in Spain: A Single-Center, Real-World, Exploratory Study. Cancers (Basel) 2024; 16:2068. [PMID: 38893187 PMCID: PMC11171292 DOI: 10.3390/cancers16112068] [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: 04/19/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The objective of this observational, single-center, retrospective study conducted in a Spanish tertiary hospital was to describe the real-world (RW) healthcare resource utilization (HCRU) among patients with advanced non-small-cell lung cancer (aNSCLC) who received chemotherapy (CT) or immunotherapy (IT) as first and second lines of treatment. A total of 173 patients diagnosed with aNSCLC and treated between January 2016 and August 2020 were included. The standardized average costs per patient/year were EUR 40,973.2 and EUR 22,502.4 for first-line CT and IT and EUR 140,601.3 and EUR 20,175.9 for second-line CT and IT, respectively. The average annual costs per patient associated with adverse-event (AE) onset were EUR 29,939.7 and EUR 460.7 for first-line CT and IT and EUR 35,906.4 and EUR 3206.1 for second-line CT and IT, respectively. The costs associated with disease management were EUR 33,178.0 and EUR 22,448.4 for first-line CT and IT and EUR 127,134.2 and EUR 19,663.9 for second-line CT and IT, respectively. In conclusion, IT use showed a lower average annual cost per patient, which was associated with lower HCRU for both disease and AE management, compared to the use of CT. However, these results should be further confirmed in the context of the currently implemented treatment schemes, including the combination of CT with single or dual IT.
Collapse
Affiliation(s)
| | - Olga Delgado
- Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain;
| | - Angel Callejo
- OBU Medical Department, AstraZeneca Farmacéutica Spain, 28050 Madrid, Spain; (A.C.); (M.D.); (C.T.)
| | - Marta Domínguez
- OBU Medical Department, AstraZeneca Farmacéutica Spain, 28050 Madrid, Spain; (A.C.); (M.D.); (C.T.)
| | - Covadonga Torres
- OBU Medical Department, AstraZeneca Farmacéutica Spain, 28050 Madrid, Spain; (A.C.); (M.D.); (C.T.)
| |
Collapse
|
20
|
Al-Husinat L, Azzam S, Al Sharie S, Al Sharie AH, Battaglini D, Robba C, Marini JJ, Thornton LT, Cruz FF, Silva PL, Rocco PRM. Effects of mechanical ventilation on the interstitial extracellular matrix in healthy lungs and lungs affected by acute respiratory distress syndrome: a narrative review. Crit Care 2024; 28:165. [PMID: 38750543 PMCID: PMC11094887 DOI: 10.1186/s13054-024-04942-y] [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: 01/17/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Mechanical ventilation, a lifesaving intervention in critical care, can lead to damage in the extracellular matrix (ECM), triggering inflammation and ventilator-induced lung injury (VILI), particularly in conditions such as acute respiratory distress syndrome (ARDS). This review discusses the detailed structure of the ECM in healthy and ARDS-affected lungs under mechanical ventilation, aiming to bridge the gap between experimental insights and clinical practice by offering a thorough understanding of lung ECM organization and the dynamics of its alteration during mechanical ventilation. MAIN TEXT Focusing on the clinical implications, we explore the potential of precise interventions targeting the ECM and cellular signaling pathways to mitigate lung damage, reduce inflammation, and ultimately improve outcomes for critically ill patients. By analyzing a range of experimental studies and clinical papers, particular attention is paid to the roles of matrix metalloproteinases (MMPs), integrins, and other molecules in ECM damage and VILI. This synthesis not only sheds light on the structural changes induced by mechanical stress but also underscores the importance of cellular responses such as inflammation, fibrosis, and excessive activation of MMPs. CONCLUSIONS This review emphasizes the significance of mechanical cues transduced by integrins and their impact on cellular behavior during ventilation, offering insights into the complex interactions between mechanical ventilation, ECM damage, and cellular signaling. By understanding these mechanisms, healthcare professionals in critical care can anticipate the consequences of mechanical ventilation and use targeted strategies to prevent or minimize ECM damage, ultimately leading to better patient management and outcomes in critical care settings.
Collapse
Affiliation(s)
- Lou'i Al-Husinat
- Department of Clinical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Saif Azzam
- Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | | | - Ahmed H Al Sharie
- Department of Pathology and Microbiology, Jordan University of Science and Technology, Irbid, Jordan
| | - Denise Battaglini
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Robba
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Dipartimento di Scienze Chirurgiche e Diagnostiche, Università Degli Studi di Genova, Genoa, Italy
| | - John J Marini
- Department of Pulmonary and Critical Care Medicine, University of Minnesota, Minneapolis, St Paul, MN, USA
| | - Lauren T Thornton
- Department of Pulmonary and Critical Care Medicine, University of Minnesota, Minneapolis, St Paul, MN, USA
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
21
|
Helgadottir H, Matikas A, Fernebro J, Frödin JE, Ekman S, Rodriguez-Wallberg KA. Fertility and reproductive concerns related to the new generation of cancer drugs and the clinical implication for young individuals undergoing treatments for solid tumors. Eur J Cancer 2024; 202:114010. [PMID: 38520926 DOI: 10.1016/j.ejca.2024.114010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
The treatment landscape of solid tumors has changed markedly in the last years. Molecularly targeted treatments and immunotherapies have been implemented and have, in many cancers, lowered the risk of relapse and prolonged survival. Patients with tumors harboring specific targetable molecular alterations or mutations are often of a younger age, and hence future fertility and family building can be important concerns in this group. However, there are great uncertainties regarding the effect of the new drugs on reproductive functions, including fertility, pregnancy and lactation and how young patients with cancers, both women and men should be advised. The goal with this review is to gather the current knowledge regarding oncofertility and the different novel therapies, including immune checkpoint inhibitors, antibody-drug conjugates, small molecules and monoclonal antibody targeted therapies. The specific circumstances and reproductive concerns in different patient groups where novel treatments have been broadly introduced are also discussed, including those with melanoma, lung, breast, colorectal and gynecological cancers. It is clear, that more awareness is needed regarding potential drug toxicity on reproductive tissues, and it is of essence that individuals are informed based on current expertise and on available fertility preservation methods.
Collapse
Affiliation(s)
- Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Skin Cancer Centrum, Theme Cancer, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| | - Alexios Matikas
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Division of Gynecological Cancer, Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Jan-Erik Frödin
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Division of Gastrointestinal Oncology, Department of Upper abdomen, Karolinska University Hospital, Sweden
| | - Simon Ekman
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
22
|
Oskarsdottir GN, Lampa E, Berglund A, Rosengren L, Ulvestad M, Boros M, Daumont MJ, Rault C, Emanuel G, Leal C, Schoemaker MJ, Wagenius G. Real-World Treatment Patterns and Survival Outcomes for Patients with Non-Metastatic Non-Small-Cell Lung Cancer in Sweden: A Nationwide Registry Analysis from the I-O Optimise Initiative. Cancers (Basel) 2024; 16:1655. [PMID: 38730607 PMCID: PMC11083854 DOI: 10.3390/cancers16091655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide, with ~40-50% of patients diagnosed with non-metastatic disease (stages IA-IIIC). The treatment landscape is evolving rapidly as immunotherapies and targeted therapy are introduced in the non-metastatic setting, creating a need to assess patient outcomes prior to their introduction. This real-world study using Swedish National Lung Cancer Registry data examined outcomes (overall survival (OS) and time to next treatment or death (TTNTD)) and treatment patterns for adults diagnosed with non-metastatic NSCLC. Baseline characteristics and OS from diagnosis were described for all patients; OS, treatment patterns, and TTNTD from treatment start were described for the treatment subgroup (patients diagnosed from 2014 onwards), stratified by disease stage and initial treatment. OS and TTNTD were described using the Kaplan-Meier estimator. The overall population (2008-2019) included 17,433 patients; the treatment subgroup included 5147 patients. Median OS (interquartile range) overall ranged from 83.3 (31.6-165.3) months (stage I patients) to 10.4 (4.3-24.2) months (stage IIIB patients). Among the treatment subgroup, median OS and TTNTD were longest among patients receiving surgery versus other anticancer treatments. These findings provide a baseline upon which to evaluate the epidemiology of non-metastatic NSCLC as newer treatments are introduced.
Collapse
Affiliation(s)
- Gudrun N. Oskarsdottir
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, V/O Hjärt- och Lungmedicin, 222 42 Lund, Sweden
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, 22 381 Lund, Sweden
| | - Erik Lampa
- Epistat AB, 753 30 Uppsala, Sweden; (E.L.); (A.B.)
| | | | - Linda Rosengren
- Medical Department Sweden, Bristol Myers Squibb, Hemvärnsgatan 9, 171 23 Solna, Sweden
| | - Maria Ulvestad
- Medical Department Nordics, Bristol Myers Squibb, Lysaker Torg 35, 1366 Lysaker, Norway;
| | - Miklos Boros
- Department of Cardiothoracic Surgery, Linköping University Hospital, 581 85 Linköping, Sweden;
| | - Melinda J. Daumont
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, 1420 Braine-L’Alleud, Belgium;
| | | | - Gabrielle Emanuel
- Real-World Data Analytics Markets, Bristol Myers Squibb, Uxbridge Business Park, Sanderson Road, Uxbridge UB8 1DH, UK;
| | | | | | - Gunnar Wagenius
- Department of Oncology-Pathology, Karolinska Institute, 141 86 Stockholm, Sweden;
- Thoracic Oncology Centre, The Cancer Theme, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
23
|
Ten Berge H, Willems B, Pan X, Dvortsin E, Aerts J, Postma MJ, Prokop M, van den Heuvel MM. Cost-effectiveness analysis of a lung cancer screening program in the netherlands: a simulation based on NELSON and NLST study outcomes. J Med Econ 2024; 27:1197-1211. [PMID: 39291295 DOI: 10.1080/13696998.2024.2404359] [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: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND In the Netherlands, lung cancer is the leading cause of cancer-related death, accounting for more than 10,000 annual deaths. Lung cancer screening (LCS) studies using low-dose computed tomography (LDCT) have demonstrated that early detection reduces lung cancer mortality. However, no LCS program has been implemented yet in the Netherlands. A national LCS program has the potential to enhance the health outcomes for lung cancer patients in the Netherlands. OBJECTIVE AND METHODS This study evaluates the cost-effectiveness of LCS compared to no-screening in the Netherlands, by simulating the screening outcomes based on data from NEderlands-Leuvens Longkanker Screenings ONderzoek (NELSON) and National Lung Screening Trial (NLST). We simulated annual screening up to 74 years of age, using inclusion criteria from the respective studies. A decision tree and Markov model was used to predict the incremental costs, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratio (ICERs) for the screening population. The analysis used a lifetime horizon and a societal perspective. RESULTS Compared to no-screening, LCS resulted in an ICER of €5,169 per QALY for the NELSON simulation, and an ICER of €17,119 per QALY for the NLST simulation. The screening costs were highly impactful for the cost-effectiveness. The most influential parameter was the CT scan cost. Cost reduction for CT from €201 to €101 per scan would reduce the ICER to €2,335 using NELSON criteria. Additionally, LCS could prevent 15,115 and 12,611 premature lung cancer deaths, accompanied by 1.66 and 1.31 QALYs gained per lung cancer case for the NELSON and NLST simulations, respectively. CONCLUSION LCS was estimated to be cost-effective in the Netherlands for both simulations at a willingness-to-pay threshold of €20,000 per QALY. Using the NELSON criteria, less than €5,500 per QALY had to be spent. Lowering the cost per CT exam would lead to a further reduction of this amount.
Collapse
Affiliation(s)
- Hilde Ten Berge
- Institute for Diagnostic Accuracy, Groningen, The Netherlands
| | - Bo Willems
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- AstraZeneca, Oncology Business Unit, The Netherlands
| | - Xuanqi Pan
- Institute for Diagnostic Accuracy, Groningen, The Netherlands
- Unit of Global Health, Faculty of Medical Sciences, University of Groningen, Groningen, The Netherlands
| | - Evgeni Dvortsin
- Institute for Diagnostic Accuracy, Groningen, The Netherlands
| | - Joachim Aerts
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maarten J Postma
- Unit of Global Health, Faculty of Medical Sciences, University of Groningen, Groningen, The Netherlands
- Department of Economics, Econometrics & Finance, Faculty of Economics & Business, University of Groningen, Groningen, The Netherlands
| | - Mathias Prokop
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michel M van den Heuvel
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Cong Z, Zhang X, Lv Z, Jiang J, Wang L, Li J, Wang J, Zhao J. Transcriptome Analysis of the Inhibitory Effects of 20(S)-Protopanaxadiol on NCI-H1299 Non-Small Cell Lung Cancer Cells. Molecules 2023; 28:5746. [PMID: 37570716 PMCID: PMC10421167 DOI: 10.3390/molecules28155746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer seriously threatens human health. To explore the molecular mechanism of 20(S)-Protopanaxadiol (PPD) on human non-small cell lung cancer cells, we investigated the transcriptional profile of PPD-treated NCI-H1299 cells. Cell proliferation, cell cycle, and apoptosis were detected using cell counting kit-8 and flow cytometry, respectively. Differentially expressed genes (DEGs) between PPD-treated and untreated cells were determined using RNA sequencing and bioinformatic analysis. Protein phosphorylation was detected using Western blotting. Data of mRNA expression profiles of lung cancer were from The Cancer Genome Atlas (TCGA) and analyzed using R software version 4.3.1. PPD showed an inhibitory effect on the proliferation of NCI-H1299 cells and induced apoptosis. There were 938 upregulated genes and 466 downregulated genes in PPD-treated cells, and DEGs were primarily enriched in the MAPK signaling pathway. The detection of phosphorylation revealed that the phosphorylation of ERK and p38 MAPK was significantly reduced in PPD-treated cells. Further comparison of PPD-regulated DEGs with clinical data of lung adenocarcinoma demonstrated that most downregulated genes in tumor tissues were upregulated in PPD-treated cells or vice versa. Two PPD-downregulated genes HSPA2 and EFNA2 were associated with patients' overall survival. Therefore, PPD could inhibit NCI-H1299 cells by affecting gene expression and regulating ERK and p38 MAPK pathways.
Collapse
Affiliation(s)
- Zhongyi Cong
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Xinmin Zhang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Zeqi Lv
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Jingyuan Jiang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Lei Wang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Jiapeng Li
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Jie Wang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Fujin Road 1266, Changchun 130021, China; (Z.C.); (X.Z.); (Z.L.); (J.J.); (L.W.); (J.L.); (J.W.)
| | - Jianjun Zhao
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Xiantai Street 126, Changchun 130033, China
| |
Collapse
|