1
|
Sheng Z, Ji S, Chen Y, Mi Z, Yu H, Zhang L, Wan S, Song N, Shen Z, Zhang P. Machine learning algorithms integrating positron emission tomography/computed tomography features to predict pathological complete response after neoadjuvant chemoimmunotherapy in lung cancer. Eur J Cardiothorac Surg 2025; 67:ezaf132. [PMID: 40221851 DOI: 10.1093/ejcts/ezaf132] [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: 12/20/2024] [Revised: 03/23/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025] Open
Abstract
OBJECTIVES Reliable methods for predicting pathological complete response (pCR) in non-small cell lung cancer (NSCLC) patients undergoing neoadjuvant chemoimmunotherapy are still under exploration. Although Fluorine-18 fluorodeoxyglucose-positron emission tomography/computed tomography (18F-FDG PET/CT) features reflect tumour response, their utility in predicting pCR remains controversial. METHODS This retrospective analysis included NSCLC patients who received neoadjuvant chemoimmunotherapy followed by 18F-FDG PET/CT imaging at Shanghai Pulmonary Hospital from October 2019 to August 2024. Eligible patients were randomly divided into training and validation cohort at a 7:3 ratio. Relevant 18F-FDG PET/CT features were evaluated as individual predictors and incorporated into 5 machine learning (ML) models. Model performance was assessed using the area under the receiver operating characteristic curve (AUC), and Shapley additive explanation was applied for model interpretation. RESULTS A total of 205 patients were included, with 91 (44.4%) achieving pCR. Post-treatment tumour maximum standardized uptake value (SUVmax) demonstrated the highest predictive performance among individual predictors, achieving an AUC of 0.72 (95% CI 0.65-0.79), while ΔT SUVmax achieved an AUC of 0.65 (95% CI 0.53-0.77). The Light Gradient Boosting Machine algorithm outperformed other models and individual predictors, achieving an average AUC of 0.87 (95% CI 0.78-0.97) in training cohort and 0.83 (95% CI 0.72-0.94) in validation cohort. Shapley additive explanation analysis identified post-treatment tumour SUVmax and post-treatment nodal volume as key contributors. CONCLUSIONS This ML models offer a non-invasive and effective approach for predicting pCR after neoadjuvant chemoimmunotherapy in NSCLC.
Collapse
Affiliation(s)
- Zhenxin Sheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuyu Ji
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yancheng Chen
- Information Service Department, Orient Overseas Container Line Limited, Shanghai, China
| | - Zirong Mi
- Department of Thoracic Surgery, Medical College of Shihezi University, Shihezi, China
| | - Huansha Yu
- Experimental Animal Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lele Zhang
- Department of Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyue Wan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Nan Song
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ziyun Shen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Gray JE, Salomonsen RJ, Diaz Perez I, Wang A, Cai L, Wetherill G, Xiao Y, Fielden C, Georgoulia N. Real-World Clinical Characteristics, Treatment Patterns, and Clinical Outcomes in US Patients with Stage I-III Resected NSCLC Without Known EGFR Mutations: The RESECT Study. Drugs Real World Outcomes 2025:10.1007/s40801-025-00487-w. [PMID: 40329045 DOI: 10.1007/s40801-025-00487-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Immunotherapy has altered the treatment landscape for resectable non-small cell lung cancer, increasing the complexity of treatment planning. Understanding treatment patterns and outcomes prior to the advent of immunotherapy can provide context for assessing the benefit of immunotherapies and other novel agents. OBJECTIVE We aimed to characterize real-world demographics, clinical characteristics, treatment patterns, and clinical outcomes of patients with early-stage non-small cell lung cancer before widespread immunotherapy use. METHODS Analyses included patients from the US CancerLinQ Discovery® database diagnosed with stage I-III non-small cell lung cancer between 2014 and 2020 without known EGFR mutations who underwent surgical resection within 140 days of diagnosis. The primary outcome was treatment patterns by disease stage. RESULTS Analyses included 3077 patients with stage I (n = 1673), II (n = 853), and III (n = 551) disease. Most (92.8%, 52.3%, and 36.5% of stage I, II, and III patients) received surgery without systemic therapy. Among stage I, II, and III patients, 7.2%, 44.8%, and 46.6% received adjuvant therapy only. Of stage II and III patients, 2.0% and 10.2% received neoadjuvant therapy only, and 0.9% and 6.7% received both (stage I patients who received neoadjuvant only or perioperative therapy were excluded because of low numbers [n = 4]). Five-year overall survival rates were 73.4%, 61.9%, and 50.5% in stage I, II, and III patients; 5-year real-world relapse-free survival rates were 35.4%, 23.1%, and 14.0%. In an exploratory multivariate analysis, neoadjuvant treatment was associated with improved overall survival and real-world relapse-free survival in stage II-III patients (stage I patients not evaluable). Adjuvant treatment was associated with improved real-world relapse-free survival, but not overall survival, in stage II-III patients. CONCLUSIONS Most patients received surgery alone, though the proportion receiving systemic treatment increased with disease stage. Modest 5-year, real-world relapse-free survival rates indicate a need for more effective neoadjuvant or adjuvant treatments in this setting.
Collapse
Affiliation(s)
- Jhanelle E Gray
- H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | | | | | | | - Ling Cai
- AstraZeneca, Gaithersburg, MD, USA
| | | | | | | | | |
Collapse
|
3
|
Madheswaran T, Chellappan DK, Lye FSN, Dua K. Recent advances in the use of liquid crystalline nanoparticles for non-small cell lung cancer treatment. Expert Opin Drug Deliv 2025; 22:615-627. [PMID: 40022612 DOI: 10.1080/17425247.2025.2474693] [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: 11/23/2024] [Revised: 01/24/2025] [Accepted: 02/27/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) continues to pose a considerable health challenge with few therapeutic alternatives. Liquid crystalline nanoparticles (LCN) are nanostructured drug delivery systems made of lipid-based amphiphilic materials that self-assemble into crystalline phases in aqueous environments. LCN have become a promising way to treat NSCLC owing to their specific properties that make them useful for targeted delivery and controlled drug release. AREAS COVERED The review provides a brief overview of the use of LCN in the treatment of NSCLC. It explores their composition, fabrication methods, and characterization processes. The article further addresses several nanoparticle-based approaches for the treatment of NSCLC. Ultimately, it underscores the promise of LCNs as a promising drug delivery system for NSCLC and discusses the obstacles and outlook in this field. EXPERT OPINION LCN represents a promising frontier in the treatment of NSCLC, offering several specific advantages over conventional therapies. Utilizing their intrinsic self-assembly characteristics, LCN provides meticulous control over drug encapsulation, release kinetics, and cellular absorption, which are crucial for improving therapy success. LCN also has the capability for co-delivery of various drugs, facilitating synergistic therapeutic benefits and addressing multidrug resistance, a prevalent issue in NSCLC treatment.
Collapse
Affiliation(s)
- Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, IMU University, Kuala Lumpur, Malaysia
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research Development and Innovation, IMU University, Kuala Lumpur, Malaysia
| | - Dinesh Kumar Chellappan
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research Development and Innovation, IMU University, Kuala Lumpur, Malaysia
- Department of Life Sciences, School of Pharmacy, IMU University, Kuala Lumpur, Malaysia
| | - Fiona Sze Nee Lye
- School of Postgraduate Studies, IMU University, Kuala Lumpur, Selangor, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
4
|
Lin GB, Chen WT, Kuo YY, Liu HH, Chen YM, Leu SJ, Chao CY. Thermal cycling‑hyperthermia sensitizes non‑small cell lung cancer A549 cells to EGFR tyrosine kinase inhibitor erlotinib. Oncol Rep 2025; 53:58. [PMID: 40183398 PMCID: PMC11976370 DOI: 10.3892/or.2025.8891] [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: 11/13/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Molecular targeted therapy has emerged as a mainstream treatment for non‑small cell lung cancer (NSCLC), the most common type of lung cancer and the leading cause of cancer‑related death in both men and women. Erlotinib (Erl), a targeted therapy inhibiting EGFR pathways, has shown notable response rate in NSCLC cells. However, limited efficacy of the treatment has been reported due to resistance among a proportion of patients with NSCLC. Therefore, sensitizers are required to potentiate the efficacy of Erl in NSCLC treatment. The present study proposed a novel thermal therapy, thermal cycling‑hyperthermia (TC‑HT), as a supplement to amplify the effects of Erl. It was demonstrated that TC‑HT reduced the half‑maximal inhibitory concentration of Erl to 0.5 µM and TC‑HT sensitized A549 NSCLC cells to Erl via the downstream EGFR signaling cascades. Furthermore, the combination treatment of Erl and TC‑HT induced G2/M cell cycle arrest and inhibition of cell proliferation and migration. In addition, by slightly raising the temperature of TC‑HT, TC‑HT treatment alone produced antineoplastic effects without damaging the normal IMR‑90 lung cells. The method presented in this study may be applicable to other combination therapies and could potentially act as a starter for anticancer treatments, with fewer side effects.
Collapse
Affiliation(s)
- Guan-Bo Lin
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
| | - Wei-Ting Chen
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
| | - Yu-Yi Kuo
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
| | - Hsu-Hsiang Liu
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 106319, Taiwan, R.O.C
| | - You-Ming Chen
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 106319, Taiwan, R.O.C
| | - Shr-Jeng Leu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Chih-Yu Chao
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 106319, Taiwan, R.O.C
| |
Collapse
|
5
|
Lin KT, Muneer G, Huang PR, Chen CS, Chen YJ. Mass Spectrometry-Based Proteomics for Next-Generation Precision Oncology. MASS SPECTROMETRY REVIEWS 2025. [PMID: 40269546 DOI: 10.1002/mas.21932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Cancer is the leading cause of death worldwide characterized by patient heterogeneity and complex tumor microenvironment. While the genomics-based testing has transformed modern medicine, the challenge of diverse clinical outcomes highlights unmet needs for precision oncology. As functional molecules regulating cellular processes, proteins hold great promise as biomarkers and drug targets. Mass spectrometry (MS)-based clinical proteomics has illuminated the molecular features of cancers and facilitated discovery of biomarkers or therapeutic targets, paving the way for innovative strategies that enhance the precision of personalized treatment. In this article, we introduced the tools and current achievements of MS-based proteomics, choice of discovery and targeted MS from discovery to validation phases, profiling sensitivity from bulk samples to single-cell level and tissue to liquid biopsy specimens, current regulatory landscape of MS-based protein laboratory-developed tests (LDTs). The challenges, success and future perspectives in translating research MS assay into clinical applications are also discussed. With well-designed validation studies to demonstrate clinical benefits and meet the regulatory requirements for both analytical and clinical performance, the future of MS-based assays is promising with numerous opportunities to improve cancer diagnosis, treatment, and monitoring.
Collapse
Affiliation(s)
- Kuen-Tyng Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Gul Muneer
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | | | - Ciao-Syuan Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
6
|
Sromek M, Głogowski M, Chechlińska M, Kulińczak M, Zajdel M, Żeber-Lubecka N, Bałabas A, Szafron ŁM, Kulecka M, Siwicki JK. Persistent and novel changes in plasma microRNA profiles in patients with non-small cell lung cancer following tumour resection. Transl Lung Cancer Res 2025; 14:677-706. [PMID: 40248723 PMCID: PMC12000959 DOI: 10.21037/tlcr-24-626] [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: 07/22/2024] [Accepted: 01/08/2025] [Indexed: 04/19/2025]
Abstract
Background Non-small cell lung cancer (NSCLC) accounts for 80% of lung cancers, the leading cause of cancer mortality. microRNAs (miRNA, miR) have emerged as important components of carcinogenesis and promising biomarkers. We aimed to analyse global plasma miRs in NSCLC patients before and at least one year after tumour resection. Methods Plasma was collected from the peripheral blood of 24 donors without cancer and of NSCLC patients before surgery (n=36) and at least 1 year after surgery (n=12). Next-generation sequencing (NGS)-based miR profiling was performed. Patients were followed-up for 4 to 12 years after surgery to assess disease recurrence. Results Untreated NSCLC patients exhibited significant changes in plasma miR levels compared to cancer-free donors (48 up- and 17 down-regulated miRs). miR profiles in patients with adenocarcinoma (ADC) (n=18) and squamous cell carcinoma (SCC) significantly differed (16 and 86 miRs up-, and 15 and 16 miRs down-regulated, respectively). A subset of pre-surgery deregulated miRs was found to be associated with recurrence (49 miRs). Six miRs were shown to have independent prognostic value. After tumour resection, some pre-surgery miR alterations returned to control levels (18 miRs), some others persisted (27 miRs), while also novel plasma miR changes emerged (75 miRs) in patients with no clinical evidence of recurrence. Conclusions Untreated NSCLC patients present deregulated plasma miRs, some of which may have a potential of prognostic markers. After tumour excision plasma miR profiles change, some miR levels normalise, some changes persist and novel miR changes are observed despite no clinical symptoms of recurrence. Plasma miR profiles in NSCLC patients may suggest systemic abnormalities predisposing to lung cancer and/or reflect a systemic response to pre-cancer/dormant cancer cells.
Collapse
Affiliation(s)
- Maria Sromek
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maciej Głogowski
- Department of Lung Cancer and Chest Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Chechlińska
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Mariusz Kulińczak
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michalina Zajdel
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Natalia Żeber-Lubecka
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Aneta Bałabas
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Łukasz M. Szafron
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Kulecka
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Jan K. Siwicki
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
7
|
Liu Y, Long J, Deng H, Chen W. Neoadjuvant immunotherapy for NSCLC: superior combination strategies, optimal treatment cycles, and predictive indicators from a Bayesian meta-analysis. Front Immunol 2025; 16:1548665. [PMID: 40213564 PMCID: PMC11983614 DOI: 10.3389/fimmu.2025.1548665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Background Neoadjuvant immune checkpoint inhibitors (ICIs) have emerged as a promising treatment strategy for resectable non-small cell lung cancer (NSCLC). However, optimal combination strategies, treatment cycles, and predictive indicators for long-term outcomes remain unclear. This study aimed to evaluate the efficacy of various neoadjuvant ICI-based therapies in resectable NSCLC, identify the optimal treatment cycles for neoadjuvant immunochemotherapy, and assess the prognostic value of pathological complete response (pCR) and major pathological response (MPR) for event-free survival (EFS). Methods A systematic literature search was conducted in PubMed, EMBASE, Cochrane CENTRAL, and Web of Science, including studies published up to October 2024. Bayesian models were used to analyze the efficacy of different ICI-based treatment combinations, assess the impact of immunochemotherapy cycles on MPR and pCR, and examine the predictive value of MPR and pCR for EFS. Results Data from 34 studies were included, consisting of 32 single-arm studies (reported in 26 papers) and 8 RCTs, involving 4,593 patients. Immunochemotherapy combined with anti-angiogenesis agents was the most effective treatment strategy, significantly improving both MPR and pCR. No significant improvement in efficacy was observed when the number of neoadjuvant immunochemotherapy cycles exceeded 3 cycles. Both MPR and pCR were strong predictors of EFS. MPR showed a stronger negative correlation with event risk compared to pCR, with a log (HR) of -2.110 (95% CI: -4.150, -0.071) for MPR, and a log (HR) of -1.665 (95% CI: -2.419, -0.992) for pCR. Conclusion Neoadjuvant immunochemotherapy combined with anti-angiogenesis agents appears to be a highly effective strategy for resectable NSCLC. Three cycles of neoadjuvant immunochemotherapy demonstrated optimal efficacy in this study. Both MPR and pCR are valuable prognostic indicators for EFS, with MPR showing a stronger predictive value. These findings offer important insights for optimizing treatment strategies and informing clinical decision-making in resectable NSCLC. Systematic review registration PROSPERO, identifier CRD42024592346.
Collapse
Affiliation(s)
- Yubingxue Liu
- Department of Health Examination and Oncology Screening Center, Chongqing University Cancer Hospital and Chongqing Cancer Institute, Chongqing, China
| | - Jianlin Long
- Department of Medical Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute, Chongqing, China
| | - Huan Deng
- Department of Medical Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute, Chongqing, China
| | - Wen Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute, Chongqing, China
| |
Collapse
|
8
|
Geng X, Jiang Y, Zeng Y, Cao W, Lu Y, Liang Y, Gu JJ, Wang B. Research trends of neoadjuvant therapy in lung cancer: a bibliometric analysis. Discov Oncol 2025; 16:321. [PMID: 40088301 PMCID: PMC11910492 DOI: 10.1007/s12672-025-02011-6] [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: 08/22/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Lung cancer is the most prevalent malignancy worldwide. Only a fraction of early-stage patients undergo radical surgery; however, many still experience recurrence and metastasis within 5 years postoperatively (approximately 30-75%). Neoadjuvant therapy has revolutionized the treatment approach for lung cancer, with a growing number of clinical trials investigating this modality. This study provides a comprehensive analysis of neoadjuvant therapy in lung cancer, intending to guide future research. METHOD To extract literature on neoadjuvant therapy for lung cancer published in the Web of Science Core Collection, spanning January 1, 2004, to December 31, 2023. Utilizing software tools including VOSviewer, CiteSpace, and GraphPad Prism to conduct bibliometric analysis and visualization studies on countries, institutions, journals, authors, co-cited references, and keywords in this field. RESULTS A sum of 6,085 research publications from 84 countries were analyzed, with the United States leading in publications on neoadjuvant therapy for lung cancer. The institution that publishes the most articles is the University of Texas System. The most published journal is Annals of Thoracic Surgery, while the most frequently co-cited journal is Journal of Clinical Oncology. Eight of the top ten co-cited references concern immune checkpoint inhibitors(ICIs). Keyword burst analysis indicates that the current research focuses and trends mainly center around four areas: ICIs, clinical trials, efficacy, and non-small cell lung cancer (NSCLC). CONCLUSIONS This is the first bibliometric study of neoadjuvant therapy in lung cancer. Over the past two decades, interest in this field has steadily increased, particularly since 2017. The United States is the largest contributor and has the highest number of publications in this field. Immune checkpoint inhibitors, clinical trials, efficacy, and NSCLC are hotspots in neoadjuvant therapy for lung cancer, both now and in the foreseeable future.
Collapse
Affiliation(s)
- Xiaoru Geng
- Clinical Medical College, Yangzhou University, Jiangsu, China
- Department of Oncology, Shuyang Hospital of Traditional Chinese Medicine, Suqian, China
| | - Youqin Jiang
- Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Yichun Zeng
- Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Wenmiao Cao
- Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Yao Lu
- Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Yichen Liang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Juan J Gu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Buhai Wang
- Clinical Medical College, Yangzhou University, Jiangsu, China.
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| |
Collapse
|
9
|
Karashima T, Takamori S, Abe M, Takumi Y, Osoegawa A, Sugio K. Safety and efficacy of neoadjuvant cisplatin + S-1 combined with radiation therapy for locally advanced non-small cell lung cancer. Surg Today 2025:10.1007/s00595-025-03019-9. [PMID: 40014076 DOI: 10.1007/s00595-025-03019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/28/2024] [Indexed: 02/28/2025]
Abstract
PURPOSE To assess the safety and efficacy of neoadjuvant chemoradiotherapy with cisplatin plus S-1 for advanced non-small cell lung cancer (NSCLC), with a focus on real-world outcomes. METHODS This retrospective study analyzed 32 patients with stage II-III NSCLC eligible for resection, who received preoperative induction therapy between January 2012 and December 2022. Specifically, 20 patients received cisplatin, S-1, and radiation therapy. RESULTS Among the 32 patients who received induction therapy, the objective response rate (ORR) was 56.2%, and surgical resection was feasible in 29 patients (90.6%). The 5 year recurrence-free survival (RFS) rate was 76.4%, and the 3- and 5 year overall survival (OS) rates were 86.2% and 82.3%, respectively. In the cisplatin + S-1 + radiation therapy group (n = 20), the ORR was 65.0%, and surgical resection was feasible in 17 patients (85.0%). The 3-year RFS and OS rates were 78.3% and 83.8%, respectively. Ef. 3 (complete pathological response) was observed in 3 patients (10.3%). No recurrences occurred in the non-adenocarcinoma subgroup (n = 6), indicating better outcomes relative to the adenocarcinoma group (5-year RFS, 100% vs. 61.4%; p = 0.07). CONCLUSIONS Induction therapy, particularly with cisplatin + S-1 + radiation was associated with promising RFS and OS in locally advanced NSCLC, with favorable tolerability and effectiveness.
Collapse
Affiliation(s)
- Takashi Karashima
- Department of Thoracic and Breast Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu, 879-5593, Japan
| | - Shinkichi Takamori
- Department of Thoracic and Breast Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu, 879-5593, Japan
| | - Miyuki Abe
- Department of Thoracic and Breast Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu, 879-5593, Japan
| | - Yohei Takumi
- Department of Thoracic and Breast Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu, 879-5593, Japan
| | - Atsushi Osoegawa
- Department of Thoracic and Breast Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu, 879-5593, Japan.
| | - Kenji Sugio
- Department of Thoracic and Breast Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu, 879-5593, Japan
| |
Collapse
|
10
|
Tahayneh K, Idkedek M, Abu Akar F. NSCLC: Current Evidence on Its Pathogenesis, Integrated Treatment, and Future Perspectives. J Clin Med 2025; 14:1025. [PMID: 39941694 PMCID: PMC11818267 DOI: 10.3390/jcm14031025] [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: 07/30/2024] [Revised: 01/11/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Non-small cell lung carcinoma (NSCLC) comprises the majority of lung cancer cases, characterized by a complex interplay of genetic alterations, environmental factors, and molecular pathways contributing to its pathogenesis. This article highlights the multifaceted pathogenesis of NSCLC and discusses screening and integrated strategies for current treatment options. NSCLC is an evolving field with various aspects including the underlying molecular alterations, oncogenic driver mutations, and immune microenvironment interactions that influence tumor progression and response to therapy. Surgical treatment remains the most applicable curative option, especially in the early stages of the disease, adjuvant chemotherapy may add benefits to previously resected patients. Combined Radio-chemotherapy can also be used for palliative purposes. There are various future perspectives and advancing horizons in NSCLC management, encompassing novel therapeutic modalities and their applications, such as CAR-T cell therapy, antibody-drug conjugates, and gene therapies. On the other hand, it's crucial to highlight the efficacy of innovative modalities of Immunotherapy and immune checkpoint inhibitors that are nowadays widely used in treatment of NSCLC. Moreover, the latest advancements in molecular profiling techniques and the development of targeted therapies designed for specific molecular alterations in NSCLC play a significant role in its treatment. In conclusion, personalized approaches are a cornerstone of successful treatment, and they are based on a patient's unique molecular profile, tumor characteristics, and host factors. Entitling the concept of individualized treatment strategies requires proper patient selection, taking into consideration mechanisms of resistance, and investigating potential combination therapies, to achieve the optimal impact on long-term survival.
Collapse
Affiliation(s)
- Kareem Tahayneh
- Faculty of Medicine, Al-Quds University, East Jerusalem 20002, Palestine;
| | - Mayar Idkedek
- Faculty of Medicine, Al-Quds University, East Jerusalem 20002, Palestine;
| | - Firas Abu Akar
- Department of General Surgery, Faculty of Medicine, Al-Quds University, East Jerusalem 20002, Palestine
- Department of Thoracic Surgery, The Edith Wolfson Medical Center, Holon 58100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
11
|
Wang L, Zheng G, Hu Y, Maolan A, Luo Y, Li Y, Liu R. Comparative efficacy and safety of first-line neoadjuvant therapy for early-stage non-small cell lung cancer based on immune checkpoint inhibitor therapy: a systematic review and network meta-analysis. BMC Pulm Med 2025; 25:49. [PMID: 39885491 PMCID: PMC11781062 DOI: 10.1186/s12890-025-03479-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
INTRODUCTION Although there are a number of neoadjuvant immunotherapy combinations that can be applied to the treatment of perioperative non-small cell lung cancer patients, the optimal treatment combination strategy has not yet been determined. METHODS We searched PubMed, EMBASE, Cochrane Library, ClinicalTrials.go and randomised controlled trials (RCTs) from major international conferences for literature related to neoadjuvant immunotherapy combinations published as first-line treatment options for non-small cell lung cancer from the start of the library to 20 February 2024, and performed a systematic review and network meta-analysis. RESULTS We analyzed nine studies involving 3431 patients, including eight perioperative neoadjuvant immunotherapy combinations for non-small cell lung cancer. For patients without programmed death-ligand 1(PD-L1) selection, Toripalimab plus chemotherapy provided the best Pathological complete response (PCR) benefit (OR = 32.89,95% CI:7.88-137.32), best Major Pathological response (MPR) benefit (OR = 10.25, 95% CI: 5.81-18.10) and best Event-free survival (EFS) benefit (HR = 0.40,95% CI: 0.28-0.57). Nivolumab plus chemotherapy provided the best surgical resection rate (OR = 1.71, 95% CI:0.87-3.40) and pembrolizumab plus chemotherapy provided the best R0 surgical resection rate (OR = 2.20, 95% CI:1.28-3.79). In contrast, the combination of ipilimumab, nivolumab and chemotherapy, and the combination of toripalimab and chemotherapy were associated with the lowest incidence of adverse events of grade 3 or above during neoadjuvant therapy. CONCLUSIONS Our findings suggest that: Toripalimab plus chemotherapy showed better neoadjuvant efficacy and may have an overall survival benefit, but also increased the incidence of serious adverse events during neoadjuvant therapy.
Collapse
Affiliation(s)
- Linfeng Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Guangda Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yue Hu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Ayidana Maolan
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yue Luo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yue Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Rui Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
12
|
Shigenobu T, Taniguchi Y, Suzuki T, Tabuchi Y, Sato M, Odagiri K, Nakamura Y, Shimokawa T, Okamoto H, Yoshizu A. Surgery versus concurrent chemoradiotherapy for stage III non-small cell lung cancer: a retrospective study with propensity score matching. BMC Cancer 2025; 25:121. [PMID: 39844071 PMCID: PMC11753074 DOI: 10.1186/s12885-025-13550-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND No study has directly compared the outcomes of surgery and concurrent chemoradiotherapy (cCRT) in patients with stage III non-small cell lung cancer (NSCLC) to date. This study aimed to compare the treatment efficacy of complete resection and definitive cCRT. METHODS Patients were recruited in this retrospective study from Yokohama Municipal Citizens' Hospital between January 2013 and December 2022. We analyzed patients with pathological stage III NSCLC who underwent complete surgical resection and those with clinical stage III NSCLC who underwent definitive cCRT. Propensity score matching was performed to balance baseline clinicopathological factors, and the prognoses of patients in each treatment group were examined using Cox proportional hazards regression. RESULTS Of the 923 patients with NSCLC who underwent surgery, 97 with pathologic stage III NSCLC underwent complete resection (surgery group) and 125 with clinical stage III NSCLC underwent cCRT (cCRT group), of whom 54 (43.2%) received consolidation therapy with durvalumab. Overall survival (OS) was significantly higher in the surgery group than in the cCRT group (5-year OS: 60.5% versus 43.0%), hazard ratio [HR] = 0.585, 95% confidence interval [CI]: 0.390-0.877, p = 0.010). However, no significant difference in OS was found between the two groups after propensity score matching (5-year OS: 59.8% versus 48.1%, HR = 0.728, 95% CI: 0.416-1.277, p = 0.268). CONCLUSIONS The outcomes of the surgery and cCRT groups did not significantly differ in the treatment of stage III NSCLC. Appropriate evaluation of the treatment required should be reviewed on a case-by-case basis.
Collapse
Affiliation(s)
- Takao Shigenobu
- Department of Thoracic Surgery, Yokohama Municipal Citizen's Hospital, 1-1, Mitsuzawanishimachi, Kanagawa-ku, Yokohama City, Kanagawa Prefecture, 221-0855, Japan.
| | - Yuri Taniguchi
- Department of Respiratory Medicine, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Takahiro Suzuki
- Department of Thoracic Surgery, Yokohama Municipal Citizen's Hospital, 1-1, Mitsuzawanishimachi, Kanagawa-ku, Yokohama City, Kanagawa Prefecture, 221-0855, Japan
| | - Yuya Tabuchi
- Department of Radiation Oncology, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Mizuki Sato
- Department of Radiation Oncology, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Kazumasa Odagiri
- Department of Radiation Oncology, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Yukiko Nakamura
- Department of Respiratory Medicine, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Tsuneo Shimokawa
- Department of Respiratory Medicine, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Hiroaki Okamoto
- Department of Respiratory Medicine, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Akira Yoshizu
- Department of Thoracic Surgery, Yokohama Municipal Citizen's Hospital, 1-1, Mitsuzawanishimachi, Kanagawa-ku, Yokohama City, Kanagawa Prefecture, 221-0855, Japan
| |
Collapse
|
13
|
Kashyap P, Raj KV, Sharma J, Dutt N, Yadav P. Classification of NSCLC subtypes using lung microbiome from resected tissue based on machine learning methods. NPJ Syst Biol Appl 2025; 11:11. [PMID: 39824879 PMCID: PMC11742043 DOI: 10.1038/s41540-025-00491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025] Open
Abstract
Classification of adenocarcinoma (AC) and squamous cell carcinoma (SCC) poses significant challenges for cytopathologists, often necessitating clinical tests and biopsies that delay treatment initiation. To address this, we developed a machine learning-based approach utilizing resected lung-tissue microbiome of AC and SCC patients for subtype classification. Differentially enriched taxa were identified using LEfSe, revealing ten potential microbial markers. Linear discriminant analysis (LDA) was subsequently applied to enhance inter-class separability. Next, benchmarking was performed across six different supervised-classification algorithms viz. logistic-regression, naïve-bayes, random-forest, extreme-gradient-boost (XGBoost), k-nearest neighbor, and deep neural network. Noteworthy, XGBoost, with an accuracy of 76.25%, and AUROC (area-under-receiver-operating-characteristic) of 0.81 with 69% specificity and 76% sensitivity, outperform the other five classification algorithms using LDA-transformed features. Validation on an independent dataset confirmed its robustness with an AUROC of 0.71, with minimal false positives and negatives. This study is the first to classify AC and SCC subtypes using lung-tissue microbiome.
Collapse
Affiliation(s)
- Pragya Kashyap
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Kalbhavi Vadhi Raj
- Department of Electrical Engineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Jyoti Sharma
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Naveen Dutt
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Pankaj Yadav
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, Rajasthan, India.
| |
Collapse
|
14
|
Reddy RA, Varshini MS, Kumar RS. Matrix Metalloproteinase-2 (MMP-2): As an Essential Factor in Cancer Progression. Recent Pat Anticancer Drug Discov 2025; 20:26-44. [PMID: 37861020 PMCID: PMC11826896 DOI: 10.2174/0115748928251754230922095544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/21/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
The development of cancer has been a multistep process involving mutation, proliferation, survival, invasion, and metastasis. Of all the characteristics of cancer, metastasis is believed to be the hallmark as it is responsible for the highest number of cancer-related deaths. In connection with this, Matrix metalloproteinases (MMPs), that has a role in metastasis, are one of the novel therapeutic targets. MMPs belong to the family of zinc-dependent endopeptidases and are capable of degrading the components of the extracellular matrix (ECM). The role of MMPs in ECM remodeling includes tissue morphogenesis, uterine cycling, growth, tissue repair, and angiogenesis. During pathological conditions, MMPs play a critical role in the excessive degradation of ECM which includes arthritis, tumour invasion, tumour metastasis, and several other autoimmune disorders. Moreover, they are believed to be involved in many physiological aspects of the cell, such as proliferation, migration, differentiation, angiogenesis, and apoptosis. It is reported that dysregulation of MMP in a variety of cancer subtypes have a dual role in tumour growth and metastasis processes. Further, multiple studies suggest the therapeutic potential of targeting MMP in invading cancer. The expression of MMP-2 correlates with the clinical characteristics of cancer patients, and its expression profile is a new diagnostic and prognostic biomarker for a variety of human diseases. Hence, manipulating the expression or function of MMP-2 may be a potential treatment strategy for different diseases, including cancers. Hence, the present review discusses the therapeutic potential of targeting MMP in various types of cancers and their recent patents.
Collapse
|
15
|
Gao C, Wu L, Wu W, Huang Y, Wang X, Sun Z, Xu M, Gao C. Deep learning in pulmonary nodule detection and segmentation: a systematic review. Eur Radiol 2025; 35:255-266. [PMID: 38985185 PMCID: PMC11632000 DOI: 10.1007/s00330-024-10907-0] [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: 01/29/2024] [Revised: 04/09/2024] [Accepted: 05/10/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVES The accurate detection and precise segmentation of lung nodules on computed tomography are key prerequisites for early diagnosis and appropriate treatment of lung cancer. This study was designed to compare detection and segmentation methods for pulmonary nodules using deep-learning techniques to fill methodological gaps and biases in the existing literature. METHODS This study utilized a systematic review with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, searching PubMed, Embase, Web of Science Core Collection, and the Cochrane Library databases up to May 10, 2023. The Quality Assessment of Diagnostic Accuracy Studies 2 criteria was used to assess the risk of bias and was adjusted with the Checklist for Artificial Intelligence in Medical Imaging. The study analyzed and extracted model performance, data sources, and task-focus information. RESULTS After screening, we included nine studies meeting our inclusion criteria. These studies were published between 2019 and 2023 and predominantly used public datasets, with the Lung Image Database Consortium Image Collection and Image Database Resource Initiative and Lung Nodule Analysis 2016 being the most common. The studies focused on detection, segmentation, and other tasks, primarily utilizing Convolutional Neural Networks for model development. Performance evaluation covered multiple metrics, including sensitivity and the Dice coefficient. CONCLUSIONS This study highlights the potential power of deep learning in lung nodule detection and segmentation. It underscores the importance of standardized data processing, code and data sharing, the value of external test datasets, and the need to balance model complexity and efficiency in future research. CLINICAL RELEVANCE STATEMENT Deep learning demonstrates significant promise in autonomously detecting and segmenting pulmonary nodules. Future research should address methodological shortcomings and variability to enhance its clinical utility. KEY POINTS Deep learning shows potential in the detection and segmentation of pulmonary nodules. There are methodological gaps and biases present in the existing literature. Factors such as external validation and transparency affect the clinical application.
Collapse
Affiliation(s)
- Chuan Gao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Linyu Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yichao Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhichao Sun
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Maosheng Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Chen Gao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
16
|
Schneider JL, Han S, Nabel CS. Fuel for thought: targeting metabolism in lung cancer. Transl Lung Cancer Res 2024; 13:3692-3717. [PMID: 39830762 PMCID: PMC11736591 DOI: 10.21037/tlcr-24-662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
For over a century, we have appreciated that the biochemical processes through which micro- and macronutrients are anabolized and catabolized-collectively referred to as "cellular metabolism"-are reprogrammed in malignancies. Cancer cells in lung tumors rewire pathways of nutrient acquisition and metabolism to meet the bioenergetic demands for unchecked proliferation. Advances in precision medicine have ushered in routine genotyping of patient lung tumors, enabling a deeper understanding of the contribution of altered metabolism to tumor biology and patient outcomes. This paradigm shift in thoracic oncology has spawned a new enthusiasm for dissecting oncogenotype-specific metabolic phenotypes and creates opportunity for selective targeting of essential tumor metabolic pathways. In this review, we discuss metabolic states across histologic and molecular subtypes of lung cancers and the additional changes in tumor metabolic pathways that occur during acquired therapeutic resistance. We summarize the clinical investigation of metabolism-specific therapies, addressing successes and limitations to guide the evaluation of these novel strategies in the clinic. Beyond changes in tumor metabolism, we also highlight how non-cellular autonomous processes merit particular consideration when manipulating metabolic processes systemically, such as efforts to disentangle how lung tumor cells influence immunometabolism. As the future of metabolic therapeutics hinges on use of models that faithfully recapitulate metabolic rewiring in lung cancer, we also discuss best practices for harmonizing workflows to capture patient specimens for translational metabolic analyses.
Collapse
Affiliation(s)
- Jaime L. Schneider
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Christopher S. Nabel
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
17
|
Li C, He A, Hu J, Xia Y, He C, Zhuang W. Mapping the evolution and frontiers of Translational Lung Cancer Research: a bibliometric analysis and literature review. Transl Lung Cancer Res 2024; 13:3764-3777. [PMID: 39830776 PMCID: PMC11736581 DOI: 10.21037/tlcr-24-653] [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: 07/28/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025]
Abstract
Background and Objective While bibliometric studies of single journals have been conducted, bibliometric mapping has not yet been used to analyze the literature published by the Translational Lung Cancer Research (TLCR). This study aimed to comprehensively review all publications of TLCR from its inception to 2024 and provide a detailed overview of its main publication characteristics. Methods This study analyzed publications from TLCR spanning 2012 to 2024 using CiteSpace, VOSviewer, and the 'Bibliometrix' package in R. Descriptive bibliometric methods were employed to examine the trends and dynamics in TLCR literature, identifying leading authors, institutions, and countries in terms of publication output. Furthermore, bibliometric maps were generated to visualize key research topics and terms, highlighting their evolution over time. Key Content and Findings The analysis included 2,032 publications in TLCR from 2012 to 2023 and 121 publications in 2024. The study revealed a positive trend in literature production, although there has been a slight recent decline in the number of articles published in the TLCR. China emerged as the most productive country (n=587), with Shanghai Jiao Tong University being the most productive institution (n=127). Jianxing He from the First Affiliated Hospital of Guangzhou Medical University was identified as the most prolific author (n=75). The top ten most cited articles primarily address treatment strategies, recurrence, immune-related toxicities, global trends in mortality, and mechanisms of resistance, reflecting the broad scope and critical importance of ongoing research in lung cancer. Research published in TLCR predominantly targeted old adults with non-small cell lung cancer (n=879), with significant emphasis on overall survival (n=507), cancer staging (n=406), and cancer immunotherapy. Conclusions This study reviewed TLCR publications from 2012 to 2024, identifying key trends, top contributors, and research focuses. Future research directions in TLCR might focus on first-line treatment, ensartinib, and advanced data analysis methods such as the Kyoto Encyclopedia of Genes and Genomes (KEGG) to revolutionize lung cancer research and practice. In conclusion, this study underscores TLCR's significant contributions to lung cancer research and provides valuable insights into its evolution and future directions.
Collapse
Affiliation(s)
- Chong Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Anqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Hu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Weihua Zhuang
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Lin N, Wu J, Adams S, Asch S, Zeliadt S, Sox-Harris A, Han S, Backhus L. Surgeon specialty and surveillance after resection for non-small cell lung cancer. JTCVS OPEN 2024; 22:470-475. [PMID: 39780819 PMCID: PMC11704532 DOI: 10.1016/j.xjon.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 01/11/2025]
Affiliation(s)
- Nicole Lin
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford School of Medicine, Stanford, Calif
| | - Julie Wu
- Division of Medical Oncology, VA Palo Alto Healthcare System, Palo Alto, Calif
| | - Scott Adams
- Health Services Research and Development, VA Puget Sound Healthcare System, Seattle, Wash
| | - Steven Asch
- Division of Medical Oncology, VA Palo Alto Healthcare System, Palo Alto, Calif
| | - Steven Zeliadt
- Health Services Research and Development, VA Puget Sound Healthcare System, Seattle, Wash
| | - Alex Sox-Harris
- Division of Medical Oncology, VA Palo Alto Healthcare System, Palo Alto, Calif
| | - Summer Han
- Department of Neurosurgery, Stanford School of Medicine, Stanford, Calif
| | - Leah Backhus
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford School of Medicine, Stanford, Calif
- Division of Medical Oncology, VA Palo Alto Healthcare System, Palo Alto, Calif
| |
Collapse
|
19
|
Jiang Q, Wei Z, Liu P, Li Z, Jiang H, Cao Y, Zhang B, Yan Y, He Y. Global trends and research hotspots in perioperative management of lung cancer: a bibliometric analysis from 2004 to 2024. Front Immunol 2024; 15:1500686. [PMID: 39640262 PMCID: PMC11617563 DOI: 10.3389/fimmu.2024.1500686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Objective This article aims to analyze the current status and research hotspots of literature related to perioperative management of patients with Lung Cancer and provide reference for future research directions. Methods This study conducted a bibliometric analysis of research literature related to perioperative management of Lung Cancer published between 2004 and 2024, retrieved from the Web of Science database. R software and VOSviewer were used for analyzing keyword clusters and research themes, revealing trends and frontiers in this field. Results A total of 4,942 studies on perioperative management of lung cancer were included. In recent years, research in this area has shown a global upward trend, with particular focus on surgical risk assessment, complication prevention, and postoperative management. Perioperative biomarkers before and after surgery have emerged as a central focus due to their impact on diagnosis and treatment. The application of novel therapies, such as targeted drugs and immunotherapy, in perioperative management is also becoming a significant research hotspot. Additionally, China has been a leading contributor to research output in this field, demonstrating strong performance in international collaborations. Conclusion Perioperative management is a critical factor influencing the prognosis of Resectable lung cancer patients. Through a systematic analysis of the current status and research hotspots in perioperative management of lung cancer, this study provides valuable references for future clinical practice and research, particularly regarding the integration of novel therapies to optimize patient outcomes.
Collapse
Affiliation(s)
- Qinling Jiang
- Department of Oncology, Nanxishan Hospital of the Guangxi Zhuang Autonomous Region, Guilin, China
| | - Zhuheng Wei
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Pingping Liu
- Department of Pharmacy, Sanya Central Hospital,The Third People’s Hospital of Hainan Province, Sanya, Hainan, China
| | - Zonghuai Li
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Huiqin Jiang
- Department of Oncology, Nanxishan Hospital of the Guangxi Zhuang Autonomous Region, Guilin, China
| | - Yilin Cao
- Department of Oncology, Nanxishan Hospital of the Guangxi Zhuang Autonomous Region, Guilin, China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Yuanyuan Yan
- Department of Pharmacy, Sanya Central Hospital,The Third People’s Hospital of Hainan Province, Sanya, Hainan, China
| | - Yulong He
- Department of Oncology, Nanxishan Hospital of the Guangxi Zhuang Autonomous Region, Guilin, China
| |
Collapse
|
20
|
Zhang X, Xiao K, Wen Y, Wu F, Gao G, Chen L, Zhou C. Multi-omics with dynamic network biomarker algorithm prefigures organ-specific metastasis of lung adenocarcinoma. Nat Commun 2024; 15:9855. [PMID: 39543109 PMCID: PMC11564768 DOI: 10.1038/s41467-024-53849-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Efficacious strategies for early detection of lung cancer metastasis are of significance for improving the survival of lung cancer patients. Here we show the marker genes and serum secretome foreshadowing the lung cancer site-specific metastasis through dynamic network biomarker (DNB) algorithm, utilizing two clinical cohorts of four major types of lung cancer distant metastases, with single-cell RNA sequencing (scRNA-seq) of primary lesions and liquid chromatography-mass spectrometry data of sera. Also, we locate the intermediate status of cancer cells, along with its gene signatures, in each metastatic state trajectory that cancer cells at this stage still have no specific organotropism. Furthermore, an integrated neural network model based on the filtered scRNA-seq data is successfully constructed and validated to predict the metastatic state trajectory of cancer cells. Overall, our study provides an insight to locate the pre-metastasis status of lung cancer and primarily examines its clinical application value, contributing to the early detection of lung cancer metastasis in a more feasible and efficacious way.
Collapse
Affiliation(s)
- Xiaoshen Zhang
- School of Medicine, Tongji University, 200092, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China
- Department of Respiratory Medicine, Shanghai Sixth People's hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Kai Xiao
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 201100, Shanghai, China
| | - Yaokai Wen
- School of Medicine, Tongji University, 200092, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 201100, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 310024, Hangzhou, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, China.
| |
Collapse
|
21
|
Chandouri B, Naves T, Yassine M, Ikhlef L, Tricard J, Chaunavel A, Homayed Z, Pannequin J, Girard N, Durand S, Carré V, Lalloué F. Comparison of methods for cancer stem cell detection in prognosis of early stages NSCLC. Br J Cancer 2024; 131:1425-1436. [PMID: 39304747 PMCID: PMC11519646 DOI: 10.1038/s41416-024-02839-9] [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: 07/28/2023] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Despite advances in diagnosis and treatment in lung cancer, therapies still fail to improve patient management due to resistance mechanisms and relapses. As Cancer stem cells (CSCs) directly contribute to tumor growth and therapeutic resistance, their clinical detection represents a major challenge. However specific and additional CSC markers lack. Thus, our aim was to achieve selective detection of CSCs with specific glycan patterns and assess the CSCs burden to predict the risk of relapse in NSCLC tumors. METHODS The lung CSCs detection and sorting with a lectin MIX were assessed and compared to CD133 in vitro. Then, its putative role as CSC biomarker was evaluated in vivo and its clinical significance on 221 NSCLC patients. RESULTS We showed a significant CSCs enrichment in the MIX+ sorted fraction compared to CD133+ cells and confirmed its high tumorigenic capacity. The MIX prognostic value on the overall survival from early stages patients was validated suggesting its potential for detecting CSCs directly linked to tumor aggressiveness. CONCLUSION The MIX could be more relevant for detecting and sorting CSCs than CD133. Moreover, its prognosis value could enable clinicians to better classify early-stage patients at high risk of relapse in order to tailor therapeutic decisions.
Collapse
Affiliation(s)
- Boutaîna Chandouri
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France.
- Carcidiag Biotechnologies company, Guéret, France.
| | - Thomas Naves
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France
| | - May Yassine
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France
| | - Léa Ikhlef
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France
| | - Jérémy Tricard
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France
- Thoracic and Cardiovascular Surgery Department, Limoges University Hospital Center, Limoges, France
| | - Alain Chaunavel
- Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Zeinab Homayed
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Nicolas Girard
- Thorax Institute Curie Montsouris, Institut Curie, Paris, France
- UVSQ, Paris Saclay University, Versailles, France
| | - Stéphanie Durand
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France.
| | | | - Fabrice Lalloué
- UMR INSERM 1308 CAPTuR, Faculty of Medicine, University of Limoges, Limoges, France.
| |
Collapse
|
22
|
Mancini A, Orlandella FM, Vitucci D, Luciano N, Alfieri A, Orrù S, Salvatore G, Buono P. Exercise's impact on lung cancer molecular mechanisms: a current overview. Front Oncol 2024; 14:1479454. [PMID: 39555455 PMCID: PMC11563951 DOI: 10.3389/fonc.2024.1479454] [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: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Lung cancer is the major cause of cancer-related deaths worldwide with an estimated 1.8 million deaths and 2.4 million new cases in 2022. Poor cardiorespiratory fitness, dyspnea and fatigue are the common features in lung cancer patients, partially limiting the exercise prescription. Exercise improves cardiorespiratory and muscular fitness and reduces the risk of some types of cancer, including lung cancer. Recently, the American Society of Clinical Oncology has encouraged preoperative exercise for lung cancer patients. Nonetheless, only limited data, mostly obtained from mouse models of lung cancer, are available on the molecular effects of exercise in lung cancer. Thus, the present minireview aims to shed light on the molecular mechanisms induced by different type of exercise in lung cancer. In particular, the role of the exercise in tumor microenvironment remodeling, angiogenesis, gene expression, apoptosis and intermediate metabolism will be examined.
Collapse
Affiliation(s)
- Annamaria Mancini
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Francesca Maria Orlandella
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Daniela Vitucci
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Neila Luciano
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Andreina Alfieri
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Stefania Orrù
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Giuliana Salvatore
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Pasqualina Buono
- Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| |
Collapse
|
23
|
Li Y, Zhao J, Li R, Yao X, Dong X, Zhao Y, Xia Z, Xu Y, Li Y. Predicting prognosis in patients with stage IA lung adenocarcinoma with a micropapillary component using a nomogram based on computed tomography radiomics and clinicopathologic factors: a retrospective analysis. Transl Lung Cancer Res 2024; 13:2585-2602. [PMID: 39507031 PMCID: PMC11535836 DOI: 10.21037/tlcr-24-544] [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: 06/24/2024] [Accepted: 09/06/2024] [Indexed: 11/08/2024]
Abstract
Background Patients with stage IA lung adenocarcinoma (ADC) with an micropapillary (MIP) component are at a higher risk of recurrence after radical surgical resection; however, adding adjuvant chemotherapy (ACT) to their postoperative course remains controversial. This study determined the predictive factors that influence the prognosis of these patients and identified those at high risk of recurrence. Methods Between January 2012 and December 2018, 254 eligible patients with stage IA lung ADC with an MIP component were categorized into training (n=169) and validation (n=85) cohorts. Clinicopathological and radiomics features were included in univariate and multivariate analyses, and statistically significant predictors were used to develop the nomogram. Area under the curve (AUC) of receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA) were used to validate the model. The calculated risk scores for each patient were risk-stratified using the X-tile procedure, and survival analyses were performed among the different risk subgroups. Results T1c stage, MIP ≥1%, spread through air space (STAS), carcinoembryonic antigen (CEA) >5 µg/L, and radiomics features were independent predictors of overall survival (OS) and disease-free survival (DFS) in patients with lung ADC with an MIP component at stage IA. Incorporating this into the nomogram, the AUCs of the nomogram predicting 3-, 5-, and 7-year OS and DFS were 0.910, 0.914, and 0.904 and 0.868, 0.838, and 0.848, respectively, in the training cohort and 0.879, 0.895, and 0.899 and 0.817, 0.805, and 0.811, respectively, in the validation cohort, showing good differentiation. The OS and DFS survival analyses among different risk subgroups showed that the nomogram could well distinguish between low- and high-risk groups. Conclusions We developed and validated a nomogram based on clinicopathological factors and radiomics features, which can be used as a powerful tool for predicting postoperative recurrence and survival in patients with stage IA lung ADC containing an MIP component.
Collapse
Affiliation(s)
- Ying Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| | - Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| | - Ruyue Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xiujing Yao
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xue Dong
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| | - Ying Zhao
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongshuo Xia
- Department of Oncology, Zibo Central Hospital, Binzhou Medical University, Zibo, China
| | - Yali Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| | - Yintao Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
24
|
Peng J, Zhang X, Hu Y, He T, Huang J, Zhao M, Meng J. Deep learning to estimate response of concurrent chemoradiotherapy in non-small-cell lung carcinoma. J Transl Med 2024; 22:896. [PMID: 39367461 PMCID: PMC11451157 DOI: 10.1186/s12967-024-05708-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Concurrent chemoradiotherapy (CCRT) is a crucial treatment for non-small cell lung carcinoma (NSCLC). However, the use of deep learning (DL) models for predicting the response to CCRT in NSCLC remains unexplored. Therefore, we constructed a DL model for estimating the response to CCRT in NSCLC and explored the associated biological signaling pathways. METHODS Overall, 229 patients with NSCLC were recruited from six hospitals. Based on contrast-enhanced computed tomography (CT) images, a three-dimensional ResNet50 algorithm was used to develop a model and validate the performance in predicting response and prognosis. An associated analysis was conducted on CT image visualization, RNA sequencing, and single-cell sequencing. RESULTS The DL model exhibited favorable predictive performance, with an area under the curve of 0.86 (95% confidence interval [CI] 0.79-0·92) in the training cohort and 0.84 (95% CI 0.75-0.94) in the validation cohort. The DL model (low score vs. high score) was an independent predictive factor; it was significantly associated with progression-free survival and overall survival in both the training (hazard ratio [HR] = 0.54 [0.36-0.80], P = 0.002; 0.44 [0.28-0.68], P < 0.001) and validation cohorts (HR = 0.46 [0.24-0.88], P = 0.008; 0.30 [0.14-0.60], P < 0.001). The DL model was also positively related to the cell adhesion molecules, the P53 signaling pathway, and natural killer cell-mediated cytotoxicity. Single-cell analysis revealed that differentially expressed genes were enriched in different immune cells. CONCLUSION The DL model demonstrated a strong predictive ability for determining the response in patients with NSCLC undergoing CCRT. Our findings contribute to understanding the potential biological mechanisms underlying treatment responses in these patients.
Collapse
Affiliation(s)
- Jie Peng
- Department of Oncology, The Second Affiliated Hospital, Guizhou Medical University, Kaili, China.
| | - Xudong Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Hu
- Department of Oncology, Guiyang Public Health Clinical Center, Guiyang, China
| | - Tianchu He
- Department of Oncology, Qiandongnan Prefecture People's Hospital, Kaili, China
| | - Jun Huang
- Department of Oncology, Qiannan Prefecture Hospital of Traditional Chinese Medicine, Duyun, China
| | - Mingdan Zhao
- Department of Oncology, Qiannan Prefecture Hospital of Traditional Chinese Medicine, Duyun, China
| | - Jimei Meng
- Department of Oncology, Qiannan Prefecture People's Hospital, Duyun, China
| |
Collapse
|
25
|
Zhang Y, Huang W, Jiao H, Kang L. PET radiomics in lung cancer: advances and translational challenges. EJNMMI Phys 2024; 11:81. [PMID: 39361110 PMCID: PMC11450131 DOI: 10.1186/s40658-024-00685-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Radiomics is an emerging field of medical imaging that aims at improving the accuracy of diagnosis, prognosis, treatment planning and monitoring non-invasively through the automated or semi-automated quantitative analysis of high-dimensional image features. Specifically in the field of nuclear medicine, radiomics utilizes imaging methods such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) to evaluate biomarkers related to metabolism, blood flow, cellular activity and some biological pathways. Lung cancer ranks among the leading causes of cancer-related deaths globally, and radiomics analysis has shown great potential in guiding individualized therapy, assessing treatment response, and predicting clinical outcomes. In this review, we summarize the current state-of-the-art radiomics progress in lung cancer, highlighting the potential benefits and existing limitations of this approach. The radiomics workflow was introduced first including image acquisition, segmentation, feature extraction, and model building. Then the published literatures were described about radiomics-based prediction models for lung cancer diagnosis, differentiation, prognosis and efficacy evaluation. Finally, we discuss current challenges and provide insights into future directions and potential opportunities for integrating radiomics into routine clinical practice.
Collapse
Affiliation(s)
- Yongbai Zhang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Hao Jiao
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China.
| |
Collapse
|
26
|
Mayenga M, Pedroso AR, Ferreira M, Gille T, Pereira Catarata MJ, Duchemann B. The CheckMate 816 trial: a milestone in neoadjuvant chemoimmunotherapy of nonsmall cell lung cancer. Breathe (Sheff) 2024; 20:240044. [PMID: 39534491 PMCID: PMC11555582 DOI: 10.1183/20734735.0044-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 08/17/2024] [Indexed: 11/16/2024] Open
Abstract
Advancements in immunotherapy in the perioperative setting have revolutionised the treatment of resectable nonsmall cell lung cancer (NSCLC). Here we present the methodology and results of the clinical trial CheckMate 816 demonstrating the benefit of neoadjuvant therapy with nivolumab plus chemotherapy compared with chemotherapy alone. Furthermore, this article discusses the implications for future practice in resectable NSCLC and the need for future research.
Collapse
Affiliation(s)
- Marie Mayenga
- Department of Pulmonology, Foch Hospital, Suresnes, France
| | - Ana Rita Pedroso
- Pulmonology Department, Local Health Unit of Braga, Braga, Portugal
| | - Marion Ferreira
- Department of Pneumology and Respiratory Functional Exploration, University Hospital of Tours, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Thomas Gille
- Inserm UMR 1272 “Hypoxia and the Lung”, UFR Santé, Médecine, Biologie Humaine (SMBH), Université Sorbonne Paris Nord (USPN), Bobigny, France
- Department of Physiology and Functional Explorations, Avicenne University Hospital, Hôpitaux Universitaires de Paris Seine-Saint-Denis (HUPSSD), Assistance Publique - Hôpitaux de Paris (AP-HP), Bobigny, France
- Departement of Physiology and Functional Explorations, Jean Verdier University Hospital, Hôpitaux Universitaires de Paris Seine-Saint-Denis (HUPSSD), Assistance Publique - Hôpitaux de Paris (AP-HP), Bondy, France
| | - Maria Joana Pereira Catarata
- Pulmonology Department, Local Health Unit of Braga, Braga, Portugal
- Tumour and Microenvironment Interactions Group, I3S-Institute for Health Research and Innovation, University of Porto, Porto, Portugal
| | - Boris Duchemann
- Department of Pulmonary Diseases, Avicenne Hospital, Bobigny, France
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
27
|
Chen K, Wang X, Yue R, Chen W, Zhu D, Cui S, Zhang X, Jin Z, Xiao T. Efficacy and safety of immune checkpoint inhibitors as neoadjuvant therapy in perioperative patients with non-small cell lung cancer: a network meta-analysis and systematic review based on randomized controlled trials. Front Immunol 2024; 15:1432813. [PMID: 39416776 PMCID: PMC11480955 DOI: 10.3389/fimmu.2024.1432813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Background Randomized controlled trials (RCTs) have unequivocally established the therapeutic advantages of combining immune checkpoint inhibitors (ICIs) with chemotherapy in the treatment of early-stage non-small cell lung cancer (NSCLC). Presently, numerous perioperative immunotherapy regimens centered around the integration of ICIs and chemotherapy have undergone clinical trials. Nonetheless, due to the absence of direct comparative RCTs among these treatment regimens, this study aims to employ Bayesian network meta-analysis to ascertain the optimal combination of ICIs and chemotherapy. Methods A systematic literature search was conducted in PubMed, EMBASE, Cochrane Library, Web of Science databases, and major international conference publications up to April 10, 2024. This comprehensive search yielded a total of 1434 studies. Following a rigorous screening process that involved evaluating the studies for relevance, methodological quality, and alignment with our research objectives, 8 studies were carefully selected for inclusion in the final analysis. Based on these curated search results, a systematic review and network meta-analysis were conducted. Results 8 RCTs were included, encompassing 7 treatments and involving 3699 operable NSCLC patients at stages I-III. Compared to chemotherapy alone, perioperative immunotherapy demonstrated higher efficacy. The combination of toripalimab and chemotherapy showed the most significant improvement in event-free survival (EFS) (HR= 0.40; 95% CI, 0.28-0.58). The regimen that most notably enhanced overall survival (OS) was Nivolumab combined with chemotherapy (HR = 0.62; 95% CI, 0.36-1.07). In terms of pathological complete response (pCR), the combination of Toripalimab and chemotherapy exhibited the highest benefit (OR = 32.89; 95% CI, 7.88-137.32). Regarding the improvement in R0 resection, Pembrolizumab plus chemotherapy performed most prominently(OR=2.15; 95% CI, 1.30-3.56). In terms of the incidence of grade 3 or higher adverse events, durvalumab combined with chemotherapy had the lowest incidence (OR = 1.05; 95% CI, 0.79-1.38), while the incidence for other regimens was higher than chemotherapy alone. Conclusion The efficacy of perioperative immunotherapy plus chemotherapy in patients with early NSCLC is significantly improved compared to chemotherapy alone. Although there is a certain risk of adverse events, the safety is within a controllable range. After a comprehensive evaluation of five endpoints in this study, it is believed that the combination of Toripalimab or Nivolumab with chemotherapy may be the optimal immunotherapy regimen for the treatment of stage Ib-IIIb NSCLC. These findings will help guide the design of clinical treatment plans and ICIs selection. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier CRD42024536799.
Collapse
Affiliation(s)
- Kaiqi Chen
- School of Basic Medical, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinwei Wang
- School of Basic Medical, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Yue
- Department of Traditional Chinese Medicine, Chongqing Changhang Hospital, Chongqing, China
| | - Wei Chen
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Danping Zhu
- Department of Endocrinology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Shikui Cui
- Department of Endocrinology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xijian Zhang
- Department of Endocrinology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Zhao Jin
- School of Basic Medical, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tong Xiao
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Harini K, Girigoswami K, Thirumalai A, Girigoswami A. Polymer-Based Antimicrobial Peptide Mimetics for Treating Multi-drug Resistant Infections: Therapy and Toxicity Evaluation. Int J Pept Res Ther 2024; 30:64. [DOI: 10.1007/s10989-024-10648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 01/03/2025]
|
29
|
Acosta-Plasencia M, He Y, Martínez D, Orozco JP, Carrasco A, Altuna-Coy A, Yang T, Díaz T, Molins L, Ramos R, Marrades RM, Navarro A. Selection of the Most Suitable Culture Medium for Patient-Derived Lung Cancer Organoids. Cells Tissues Organs 2024; 214:114-127. [PMID: 39236699 PMCID: PMC11965841 DOI: 10.1159/000541274] [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: 04/04/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024] Open
Abstract
INTRODUCTION Patient-derived organoids have emerged as a promising in vitro model for precision medicine, particularly in cancer, but also in noncancer-related diseases. However, the optimal culture medium for culturing patient-derived lung organoids has not yet been agreed upon. This study aimed to shed light on the optimal selection of a culture media for developing studies using patient-derived lung organoids. METHODS Tumor and normal paired tissue from 71 resected non-small cell lung cancer patients were processed for organoid culture. Lung cancer organoids (LCOs) were derived from tumor tissue and normal lung organoids (LNOs) from nonneoplastic lung tissue. Three different culture media were compared: permissive culture medium (PCM), limited culture medium (LCM), and minimum basal medium (MBM). We assessed their effectiveness in establishing organoid cultures, promoting organoid growth and viability, and compared their differential phenotypic characteristics. RESULTS While PCM was associated with the highest success rate and useful for long-term expansion, MBM was the best option to avoid normal organoid overgrowth in the organoid culture. The density, size, and viability of LNOs were reduced using LCM and severely affected with MBM. LNOs cultured in PCM tend to differentiate to bronchospheres, while alveolosphere differentiation can be observed in those cultured with LCM. The morphological phenotype of LCO was influenced by the culture media of election. Mesenchymal cell overgrowth was observed when LCM was used. CONCLUSION This work highlights the importance of considering the research objectives when selecting the most suitable culture medium for growing patient-derived lung organoids. INTRODUCTION Patient-derived organoids have emerged as a promising in vitro model for precision medicine, particularly in cancer, but also in noncancer-related diseases. However, the optimal culture medium for culturing patient-derived lung organoids has not yet been agreed upon. This study aimed to shed light on the optimal selection of a culture media for developing studies using patient-derived lung organoids. METHODS Tumor and normal paired tissue from 71 resected non-small cell lung cancer patients were processed for organoid culture. Lung cancer organoids (LCOs) were derived from tumor tissue and normal lung organoids (LNOs) from nonneoplastic lung tissue. Three different culture media were compared: permissive culture medium (PCM), limited culture medium (LCM), and minimum basal medium (MBM). We assessed their effectiveness in establishing organoid cultures, promoting organoid growth and viability, and compared their differential phenotypic characteristics. RESULTS While PCM was associated with the highest success rate and useful for long-term expansion, MBM was the best option to avoid normal organoid overgrowth in the organoid culture. The density, size, and viability of LNOs were reduced using LCM and severely affected with MBM. LNOs cultured in PCM tend to differentiate to bronchospheres, while alveolosphere differentiation can be observed in those cultured with LCM. The morphological phenotype of LCO was influenced by the culture media of election. Mesenchymal cell overgrowth was observed when LCM was used. CONCLUSION This work highlights the importance of considering the research objectives when selecting the most suitable culture medium for growing patient-derived lung organoids.
Collapse
Affiliation(s)
- Melissa Acosta-Plasencia
- Molecular Oncology and Embryology Laboratory, Department of Surgery and Medical Specializations, Human Anatomy and Embryology Unit, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Yangyi He
- Molecular Oncology and Embryology Laboratory, Department of Surgery and Medical Specializations, Human Anatomy and Embryology Unit, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), Barcelona, Spain
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Daniel Martínez
- Department of Pathology, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
- Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan Pablo Orozco
- Department of Pneumology, Institut Clínic Respiratori (ICR), Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Antonio Carrasco
- Department of Pathology, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Antonio Altuna-Coy
- Molecular Oncology and Embryology Laboratory, Department of Surgery and Medical Specializations, Human Anatomy and Embryology Unit, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Tianmiao Yang
- Molecular Oncology and Embryology Laboratory, Department of Surgery and Medical Specializations, Human Anatomy and Embryology Unit, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Tania Díaz
- Molecular Oncology and Embryology Laboratory, Department of Surgery and Medical Specializations, Human Anatomy and Embryology Unit, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Laureano Molins
- Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Thoracic Surgery, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Ricard Ramos
- Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Thoracic Surgery, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Ramón M. Marrades
- Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Pneumology, Institut Clínic Respiratori (ICR), Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfons Navarro
- Molecular Oncology and Embryology Laboratory, Department of Surgery and Medical Specializations, Human Anatomy and Embryology Unit, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), Barcelona, Spain
- Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
30
|
Yildirim S, Alan O, Yuksel Yasar Z, Kaya T, Akdag G, Kinikoglu O, Gecmen GG, Yasar A, Isik D, Surmeli H, Basoglu T, Sever ON, Yildirim ME, Odabas H, Turan N. Prognostic Impact and Clinical Features of Spread through Air Spaces in Operated Lung Cancer: Real-World Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1374. [PMID: 39202654 PMCID: PMC11356374 DOI: 10.3390/medicina60081374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: Lung cancer is the leading cause of cancer-related deaths. Spread through air spaces (STAS) is an adverse prognostic factor that has become increasingly known in recent years. This study aims to investigate the impact of STAS presence on overall survival (OS) and disease-free survival (DFS) in patients with surgically resected stage IA-IIIA lung cancer and to identify clinicopathological features associated with STAS. Materials and Methods: This research involved 311 lung cancer surgery patients. The relationship between the presence of STAS in the patients' surgical pathology and OS and DFS values was examined. Clinicopathological features associated with the presence of STAS were determined. Results: There were 103 (33%) STAS-positive patients. Adenocarcinoma histological subtype, perineural invasion (PNI), and lymphovascular invasion (LVI) were significantly correlated with being STAS positive. STAS significantly predicted DFS and OS. One-year and five-year DFS rates were significantly lower in the STAS-positive group compared to the STAS-negative group (65% vs. 88%, 29% vs. 62%, respectively, p ≤ 0.001). Similarly, one-year and five-year OS rates were significantly lower in the STAS-positive group compared to the STAS-negative group (92% vs. 94%, 54% vs. 88%, respectively, p ≤ 0.001). In multivariate analysis, STAS was found to be an independent prognostic factor for both DFS and OS (HR: 3.2 (95%CI: 2.1-4.8) and 3.1 (95%CI: 1.7-5.5), p < 0.001 and <0.001, respectively). Conclusions: In our study, STAS was found to be an independent prognostic biomarker in operated stage IA-IIIA lung cancer patients. It may be a beneficial pathological biomarker in predicting the survival of patients and managing their treatments.
Collapse
Affiliation(s)
- Sedat Yildirim
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Ozkan Alan
- Division of Medical Oncology, School of Medicine, Koc University, Istanbul 34450, Turkey;
| | - Zeynep Yuksel Yasar
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Tugba Kaya
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Goncagul Akdag
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Oguzcan Kinikoglu
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Gonca Gul Gecmen
- Department of Pathology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey;
| | - Alper Yasar
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Deniz Isik
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Heves Surmeli
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Tugba Basoglu
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Ozlem Nuray Sever
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Mahmut Emre Yildirim
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Hatice Odabas
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| | - Nedim Turan
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (Z.Y.Y.); (T.K.); (G.A.); (O.K.); (A.Y.); (D.I.); (H.S.); (T.B.); (O.N.S.); (M.E.Y.); (H.O.); (N.T.)
| |
Collapse
|
31
|
Petracci E, Pasini L, Urbini M, Felip E, Stella F, Davoli F, Salvi M, Beau-Faller M, Tebaldi M, Azzali I, Canale M, Solli P, Lai G, Amat R, Carbonell C, Falcoz PE, Martinez-Marti A, Pencreach E, Delmonte A, Crinò L, Ulivi P. Circulating cell-free and extracellular vesicles-derived microRNA as prognostic biomarkers in patients with early-stage NSCLC: results from RESTING study. J Exp Clin Cancer Res 2024; 43:241. [PMID: 39169404 PMCID: PMC11340091 DOI: 10.1186/s13046-024-03156-y] [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: 04/12/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Factors to accurately stratify patients with early-stage non-small cell lung cancer (NSCLC) in different prognostic groups are still needed. This study aims to investigate 1) the prognostic potential of circulating cell-free (CF) and extracellular vesicles (EVs)-derived microRNA (miRNAs), and 2) their added value with respect to known prognostic factors (PFs). METHODS The RESTING study is a multicentre prospective observational cohort study on resected stage IA-IIIA patients with NSCLC. The primary end-point was disease-free survival (DFS), and the main analyses were carried out separately for CF- and EV-miRNAs. CF- and EV-miRNAs were isolated from plasma, and miRNA-specific libraries were prepared and sequenced. To reach the study aims, three statistical models were specified: one using the miRNA data only (Model 1); one using both miRNAs and known PFs (age, gender, and pathological stage) (Model 2), and one using the PFs alone (Model 3). Five-fold cross-validation (CV) was used to assess the predictive performance of each. Standard Cox regression and elastic net regularized Cox regression were used. RESULTS A total of 222 patients were enrolled. The median follow-up time was 26.3 (95% CI 25.4-27.6) months. From Model 1, three CF-miRNAs and 21 EV-miRNAs were associated with DFS. In Model 2, two CF-miRNAs (miR-29c-3p and miR-877-3p) and five EV-miRNAs (miR-181a-2-3p, miR-182-5p, miR-192-5p, miR-532-3p and miR-589-5p) remained associated with DFS. From pathway enrichment analysis, TGF-beta and NOTCH were the most involved pathways. CONCLUSION This study identified promising prognostic CF- and EV-miRNAs that could be used as a non-invasive, cost-effective tool to aid clinical decision-making. However, further evaluation of the obtained miRNAs in an external cohort of patients is warranted.
Collapse
Affiliation(s)
- Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Luigi Pasini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Enriqueta Felip
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Franco Stella
- Thoracic Surgery Department AUSL Romagna, Forlì, Italy
| | - Fabio Davoli
- Thoracic Surgery Department AUSL Romagna, Ravenna, Italy
| | - Maurizio Salvi
- Thoracic Surgery Department AUSL Romagna, Riccione, Italy
| | - Michele Beau-Faller
- Molecular Laboratory, University Hospital, Strasbourg University, Strasburg, France
| | - Michela Tebaldi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Azzali
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Matteo Canale
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Piergiorgio Solli
- Unit of Thoracic Surgery and Lung Transplantation, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giulia Lai
- Unit of Thoracic Surgery and Lung Transplantation, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Ramon Amat
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Pierre-Emmanuel Falcoz
- Thoracic Surgery Department, Nouvel Hôpital Civil', University Hospital, Strasburg, France
| | | | - Erwan Pencreach
- Molecular Laboratory, University Hospital, Strasbourg University, Strasburg, France
| | - Angelo Delmonte
- Oncology Department, Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST) IRCCS, Meldola, Italy
| | - Lucio Crinò
- Oncology Department, Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| |
Collapse
|
32
|
Ajuwon OR, Nsole-Biteghe FA, Ndong JD, Davids LM, Ajiboye BO, Brai B, Bamisaye FA, Falode JA, Odoh IM, Adegbite KI, Adegoke BO, Ntwasa M, Lebelo SL, Ayeleso AO. Nrf2-Mediated Antioxidant Response and Drug Efflux Transporters Upregulation as Possible Mechanisms of Resistance in Photodynamic Therapy of Cancers. Onco Targets Ther 2024; 17:605-627. [PMID: 39131905 PMCID: PMC11313505 DOI: 10.2147/ott.s457749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/08/2024] [Indexed: 08/13/2024] Open
Abstract
Photodynamic therapy (PDT) is a groundbreaking approach involving the induction of cytotoxic reactive oxygen species (ROS) within tumors through visible light activation of photosensitizers (PS) in the presence of molecular oxygen. This innovative therapy has demonstrated success in treating various cancers. While PDT proves highly effective in most solid tumors, there are indications that certain cancers exhibit resistance, and some initially responsive cancers may develop intrinsic or acquired resistance to PDT. The molecular mechanisms underlying this resistance are not fully understood. Recent evidence suggests that, akin to other traditional cancer treatments, the activation of survival pathways, such as the KEAP1/Nrf2 signaling pathway, is emerging as an important mechanism of post-PDT resistance in many cancers. This article explores the dual role of Nrf2, highlighting evidence linking aberrant Nrf2 expression to treatment resistance across a range of cancers. Additionally, it delves into the specific role of Nrf2 in the context of photodynamic therapy for cancers, emphasizing evidence that suggests Nrf2-mediated upregulation of antioxidant responses and induction of drug efflux transporters are potential mechanisms of resistance to PDT in diverse cancer types. Therefore, understanding the specific role(s) of Nrf2 in PDT resistance may pave the way for the development of more effective cancer treatments using PDT.
Collapse
Affiliation(s)
| | | | | | | | | | - Bartholomew Brai
- Department of Biochemistry, Federal University, Oye-Ekiti, Ekiti State, Nigeria
| | | | - John Adeolu Falode
- Department of Biochemistry, Federal University, Oye-Ekiti, Ekiti State, Nigeria
| | - Ikenna Maximillian Odoh
- Department of Biochemistry, Federal University, Oye-Ekiti, Ekiti State, Nigeria
- Medical Center, Federal University, Oye-Ekiti, Ekiti-State, Nigeria
| | - Kabirat Iyabode Adegbite
- Department of Environmental Health Science, College of Basic Medical and Health Sciences, Fountain University, Osogbo, Osun State, Nigeria
| | | | - Monde Ntwasa
- Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodeport, South Africa
| | - Sogolo Lucky Lebelo
- Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodeport, South Africa
| | - Ademola Olabode Ayeleso
- Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodeport, South Africa
- Biochemistry Programme, Bowen University, Iwo, Osun State, Nigeria
| |
Collapse
|
33
|
Hart M, Isuri RK, Ramos D, Osharovich SA, Rodriguez AE, Harmsen S, Dudek GC, Huck JL, Holt DE, Popov AV, Singhal S, Delikatny EJ. Non-Small Cell Lung Cancer Imaging Using a Phospholipase A2 Activatable Fluorophore. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:490-500. [PMID: 39056064 PMCID: PMC11267604 DOI: 10.1021/cbmi.4c00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 07/28/2024]
Abstract
Lung cancer, the most common cause of cancer-related death in the United States, requires advanced intraoperative detection methods to improve evaluation of surgical margins. In this study we employed DDAO-arachidonate (DDAO-A), a phospholipase A2 (PLA2) activatable fluorophore, designed for the specific optical identification of lung cancers in real-time during surgery. The in vitro fluorescence activation of DDAO-A by porcine sPLA2 was tested in various liposomal formulations, with 100 nm extruded EggPC showing the best overall characteristics. Extruded EggPC liposomes containing DDAO-A were tested for their stability under various storage conditions, demonstrating excellent stability for up to 4 weeks when stored at -20 °C or below. Cell studies using KLN 205 and LLC1 lung cancer cell lines showed DDAO-A activation was proportional to cell number. DDAO-A showed preferential activation by human recombinant cPLA2, an isoform highly specific to arachidonic acid-containing lipids, when compared to a control probe, DDAO palmitate (DDAO-P). In vivo studies with DBA/2 mice bearing KLN 205 lung tumors recapitulated these results, with preferential activation of DDAO-A relative to DDAO-P following intratumoral injection. Topical application of DDAO-A-containing liposomes to human (n = 10) and canine (n = 3) lung cancers ex vivo demonstrated the preferential activation of DDAO-A in tumor tissue relative to adjacent normal lung tissue, with fluorescent tumor-to-normal ratios (TNR) of up to 5.2:1. The combined results highlight DDAO-A as a promising candidate for clinical applications, showcasing its potential utility in intraoperative and back-table imaging and topical administration during lung cancer surgeries. By addressing the challenge of residual microscopic disease at resection margins and offering stability in liposomal formulations, DDAO-A emerges as a potentially valuable tool for advancing precision lung cancer surgery and improving curative resection rates.
Collapse
Affiliation(s)
- Michael
C. Hart
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ritesh K. Isuri
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Drew Ramos
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sofya A. Osharovich
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Andrea E. Rodriguez
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Stefan Harmsen
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Grace C. Dudek
- Department
of Biology, University of Pennsylvania, 102 Leidy Laboratories 433 S University
Ave, Philadelphia, Pennsylvania 19104, United States
| | - Jennifer L. Huck
- Department
of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David E. Holt
- Department
of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Anatoliy V. Popov
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sunil Singhal
- Department
of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Edward J. Delikatny
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
34
|
Girard N, Besada M, Rogula B, Lucherini S, Vo L, Chaudhary MA, Goring S, Lozano-Ortega G, Tran M, Varol N, Waser N, Lee JM, Spicer J. Comparative Efficacy of Neoadjuvant Nivolumab Plus Chemotherapy versus Conventional Comparator Treatments in Resectable Non-Small-Cell Lung Cancer: A Systematic Literature Review and Network Meta-Analysis. Cancers (Basel) 2024; 16:2492. [PMID: 39001554 PMCID: PMC11240383 DOI: 10.3390/cancers16132492] [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: 05/25/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND This study aimed to estimate the relative efficacy of neoadjuvant nivolumab in combination with chemotherapy (neoNIVO + CT) compared to relevant treatments amongst resectable non-metastatic non-small-cell lung cancer (rNSCLC) patients. METHODS Treatment comparisons were based on a network meta-analysis (NMA) using randomized clinical trial data identified via systematic literature review (SLR). The outcomes of interest were event-free survival (EFS) and pathological complete response (pCR). NeoNIVO + CT was compared to neoadjuvant chemotherapy (neoCT), neoadjuvant chemoradiotherapy (neoCRT), adjuvant chemotherapy (adjCT), and surgery alone (S). Due to the potential for effect modification by stage, all-stage and stage-specific networks were considered. Fixed-effect (FE) and random-effects Bayesian NMA models were run (EFS = hazard ratios [HR]; pCR = odds ratios [OR]; 95% credible intervals [CrI]). RESULTS Sixty-one RCTs were identified (base case = 9 RCTs [n = 1978 patients]). In the all-stages FE model, neoNIVO + CT had statistically significant EFS improvements relative to neoCT (HR = 0.68 [95% CrI: 0.49, 0.94]), S (0.59 [0.42, 0.82]), adjCT (0.66 [0.45, 0.96]), but not relative to neoCRT (HR = 0.77 [0.52, 1.16]). NeoNIVO + CT (5 RCTs) had statistically significant higher odds of pCR relative to neoCT (OR = 12.53 [5.60, 33.82]) and neoCRT (7.15 [2.31, 24.34]). Stage-specific model findings were consistent. CONCLUSIONS This NMA signals improved EFS and/or pCR of neoNIVO + CT relative to comparators among patients with rNSCLC.
Collapse
Affiliation(s)
- Nicolas Girard
- Department of Medical Oncology, Institut Curie, 75005 Paris, France
- Paris Saclay University, University of Versailles Saint-Quentin-en-Yvelines (UVSQ), 78000 Versailles, France
| | | | | | | | - Lien Vo
- Bristol Myers Squibb, Lawrenceville, NJ 08648, USA
| | | | | | | | - Mia Tran
- Bristol Myers Squibb, Lawrenceville, NJ 08648, USA
| | | | | | | | - Jonathan Spicer
- Department of Thoracic Surgery, McGill University Health Center, Montreal, QC H3G 1A4, Canada
| |
Collapse
|
35
|
Wilkinson AN. Feuilles de route de la survivance au cancer. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2024; 70:469-473. [PMID: 39122424 PMCID: PMC11328707 DOI: 10.46747/cfp.700708469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Affiliation(s)
- Anna N Wilkinson
- Professeure agrégée au Département de médecine familiale de l'Université d'Ottawa (Ontario), directrice du programme de troisième année de résidence MF-oncologie et directrice régionale des soins primaires en cancérologie pour la région de Champlain
| |
Collapse
|
36
|
Wilkinson AN. Cancer survivorship road map. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2024; 70:465-468. [PMID: 39122429 PMCID: PMC11328720 DOI: 10.46747/cfp.700708465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Affiliation(s)
- Anna N Wilkinson
- Associate Professor in the Department of Family Medicine at the University of Ottawa in Ontario, a family physician with the Ottawa Academic Family Health Team, a general practitioner oncologist at the Ottawa Hospital Cancer Centre, Program Director of PGY-3 FP-Oncology, and Regional Cancer Primary Care Lead for Champlain Region
| |
Collapse
|
37
|
Shigefuku S, Takahashi S, Hagiwara M, Kakihana M, Ohira T, Ikeda N. Prognostic significance of immunotherapy in postoperative recurrent non-small cell lung cancer without EGFR mutations or ALK rearrangements. J Thorac Dis 2024; 16:3583-3592. [PMID: 38983180 PMCID: PMC11228743 DOI: 10.21037/jtd-24-237] [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: 02/13/2024] [Accepted: 04/26/2024] [Indexed: 07/11/2024]
Abstract
Background Limited reports exist regarding postoperative recurrent non-small cell lung cancer (NSCLC) without major driver mutations [epidermal growth factor receptor (EGFR) mutations or anaplastic lymphoma kinase (ALK) rearrangements] treated with immune checkpoint inhibitors (ICIs) when programmed cell death ligand 1 (PD-L1) is expressed in a real-world setting. The aim of this study was to evaluate the effect of ICIs for those NSCLC. Methods We enrolled 255 patients with postoperative recurrent NSCLC lacking EGFR mutations or ALK rearrangements who underwent lobectomy or more extensive resection between 2012 and 2021. Factors associated with post-recurrence survival (PRS) were determined using the Cox proportional hazards model. PRS was analyzed using Kaplan-Meier curves and compared using the log-rank test. Results Multivariable analysis demonstrated that squamous cell carcinoma, pathological stage III, and an Eastern Cooperative Oncology Group (ECOG) performance status ≥2 were significantly associated with worse PRS. Conversely, ICI use at first line was associated with improved PRS. Patients who used ICIs during the first line and subsequent therapies had better PRS than those who received chemotherapy alone. Among patients who used ICIs, there was no significant difference in response rate at the first line, nor in PRS among those with PD-L1 expression ≥50%, 1-49%, and <1% in surgically resected specimens. Conclusions ICI use at any treatment line improved the PRS of NSCLC patients without major driver mutations, irrespective of PD-L1 expression, in a real-world setting.
Collapse
Affiliation(s)
| | - Satoshi Takahashi
- Department of Surgery, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Masaru Hagiwara
- Department of Surgery, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | | | - Tatsuo Ohira
- Department of Surgery, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Norihiko Ikeda
- Department of Surgery, Tokyo Medical University, Shinjuku, Tokyo, Japan
| |
Collapse
|
38
|
Roshan-Zamir M, Khademolhosseini A, Rajalingam K, Ghaderi A, Rajalingam R. The genomic landscape of the immune system in lung cancer: present insights and continuing investigations. Front Genet 2024; 15:1414487. [PMID: 38983267 PMCID: PMC11231382 DOI: 10.3389/fgene.2024.1414487] [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: 04/09/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Lung cancer is one of the most prevalent malignancies worldwide, contributing to over a million cancer-related deaths annually. Despite extensive research investigating the genetic factors associated with lung cancer susceptibility and prognosis, few studies have explored genetic predispositions regarding the immune system. This review discusses the most recent genomic findings related to the susceptibility to or protection against lung cancer, patient survival, and therapeutic responses. The results demonstrated the effect of immunogenetic variations in immune system-related genes associated with innate and adaptive immune responses, cytokine, and chemokine secretions, and signaling pathways. These genetic diversities may affect the crosstalk between tumor and immune cells within the tumor microenvironment, influencing cancer progression, invasion, and prognosis. Given the considerable variability in the individual immunegenomics profiles, future studies should prioritize large-scale analyses to identify potential genetic variations associated with lung cancer using highthroughput technologies across different populations. This approach will provide further information for predicting response to targeted therapy and promotes the development of new measures for individualized cancer treatment.
Collapse
Affiliation(s)
- Mina Roshan-Zamir
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Khademolhosseini
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kavi Rajalingam
- Cowell College, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Abbas Ghaderi
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
39
|
Mohamed SIA, Elsayed GH, El Shaffai A, Yahya SMM, Mettwally WSA. In-vitro study of cytotoxic and apoptotic potential of Thalassia hemprichii (Ehren.) Asch. And Enhalus acoroides (L.f.) Royle against human breast cancer cell line (MCF-7) with correlation to their chemical profile. BMC Complement Med Ther 2024; 24:244. [PMID: 38915036 PMCID: PMC11194981 DOI: 10.1186/s12906-024-04512-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 05/22/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Breast cancer is the most common type of cancer diagnosed in women. Finding novel therapeutic agents with significant cytotoxic action and minimal adverse impact on normal cells becomes crucial. Today, natural anticancer agents present an unconventional method of treating cancer, either as a curative or preventative agent, with considerable concern for marine organisms. METHODS The anticancer effect of the alcoholic extract of different Red Sea Seagrasses on MCF-7 human breast cancer cell line has been investigated. Seagrasses were collected from Wadi El Gamal, Red Sea and extracted. Qualitative HPLC analysis was performed on the extracts for the identification of their active biomarkers. This study was aimed to explore the cytotoxic impact of Thalassia hemprichii (Ehren.) and Enhalus acoroides (L.f.) Royle on MCF-7 and their mode of action. Their anti-proliferative effects on cancer cells were performed using Neutral red assay. On the other hand, their apoptotic effect and their capacity to induce cell cycle arrest were investigated by flow cytometry assay. The effect of Seagrasses on the mitochondrial membrane potential (ΔψM) was studied by using JC-1 mitochondrial membrane potential assay kit in Seagrasses treated cancer cells to Δψ Caspases 3/7activity was examined using the colorimetric method. Gene expression analysis and quantitative real time RT-PCR for the sea grasses on MCF-7 was performed. Immune-blotting technique for Bcl-2 and p53 was investigated. RESULTS HPLC analysis demonstrated that the extracts contained mainly flavonoids and polyphenols such as Caffeic acid, Chlorogenic acids, catechin and kaempferol that might be responsible for these anticancer effects. Seagrasses alcoholic crude extract markedly suppressed the growth and expansion of MCF-7 cells concentration-dependently with no toxicity against normal human skin fibroblast HSF. Thalassia hemprichii and Enhalus acoroides trigger mode of cell death primarily via apoptosis as confirmed by the flow cytometry. Additionally, they have ability to induce G0/S cell cycle arrest in MCF-7. The data showed the depletion in mitochondrial membrane potential (ΔψM) in the treated cells dose-dependently Caspases 3/7activities markedly increased following 24 h treatment. Finally, Gene expression analysis showed a marked reduction in Bcl-2, Survivin and CDC2 gene expression levels and a significant increase in the expression of p53 and CC2D1A as compared to control cells. CONCLUSION In summary, the Methanolic extract of seagrass, Thalassia hemperchii and Enhalus ocoroides are able to induce concentration-dependent cytotoxic effects in human MCF-7 cells through intrinsic pathway of apoptosis in MCF-7 cells. This study reveals the beneficial importance of sea grasses as a source of anticancer agents. Further in vivo study is recommended for the active isolated biomolecules.
Collapse
Affiliation(s)
- Shimaa I A Mohamed
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, ElBuhous St. 12622, Dokki, Dokki, Giza, 12622, Egypt.
| | - Ghada H Elsayed
- Hormones Department, Medical Research and Clinical Studies Institute and Stem Cell Lab, Centre of Excellence for Advanced Sciences, National Research Centre, ElBuhous St. 12622, Dokki, Cairo, Giza, Egypt
| | - Amgad El Shaffai
- Nature Conservation Sector, Egyptian Environmental Affairs Agency (EEAA), Ministry of Environment, Cairo, Egypt
| | - Shaymaa M M Yahya
- Hormones Department, Medical Research and Clinical Studies Institute and Stem Cell Lab, Centre of Excellence for Advanced Sciences, National Research Centre, ElBuhous St. 12622, Dokki, Cairo, Giza, Egypt
| | - Walaa S A Mettwally
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, ElBuhous St. 12622, Dokki, Dokki, Giza, 12622, Egypt
| |
Collapse
|
40
|
Xiao M, Tu L, Zhou T, He Y, Li X, Zuo Q. Predictive model based on multiple immunofluorescence quantitative analysis for pathological complete response to neoadjuvant immunochemotherapy in lung squamous cell carcinoma. Front Oncol 2024; 14:1396439. [PMID: 38887237 PMCID: PMC11180808 DOI: 10.3389/fonc.2024.1396439] [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: 03/05/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Objective This study aims to establish a prediction model for neoadjuvant immunochemotherapy (NICT) in lung squamous cell carcinoma to guide clinical treatment. Methods This retrospective study included 50 patients diagnosed with lung squamous cell carcinoma who received NICT. The patients were divided into the pathological complete response (PCR) group and the non-PCR group. HE staining and multiple immunofluorescence (mIF) techniques were utilized to analyze the differences in the immune microenvironment between these groups. LASSO regression and optimal subset regression were employed to identify the most significant variables and construct a prediction model. Results The PCR group showed higher densities of lymphocyte nuclei and karyorrhexis based on HE staining. Furthermore, based on mIF analysis, the PCR group showed higher cell densities of CD8+, PD-L1+, and CD8+PD-L1+ in the tumor region, while showing lower cell densities of CD3+Foxp3+, Foxp3+, and CD163+. Logistic univariate analysis revealed CD8+PD-L1+, PD-L1+, CD8+, CD4+LAG-3+, lymphocyte nuclei, and karyorrhexis as significant factors influencing PCR. By using diverse screening methods, the three most relevant variables (CD8+, PD-L1+, and CD8+PD-L1+ in the tumor region) were selected to establish the prediction model. The model exhibited excellent performance in both the training set (AUC=0.965) and the validation set (AUC=0.786). In the validation set, In comparison to the conventional TPS scoring criteria, the model attained superior accuracy (0.85), specificity(0.67), and sensitivity (0.92). Conclusion NICT treatment might induce anti-tumor effects by enriching immune cells and reactivating exhausted T cells. CD8+, PD-L1+, and CD8+PD-L1+ cell abundances within the tumor region have been closely associated with therapeutic efficacy. Incorporating these three variables into a predictive model allows accurate forecasting of treatment outcomes and provides a reliable basis for selecting NICT treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | - Xiaohui Li
- The Geriatric Respiratory Department, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiunan Zuo
- The Geriatric Respiratory Department, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
41
|
Suzuki J, Miyoshi T, Tane K, Onodera K, Koike Y, Sakai T, Samejima J, Aokage K, Tsuboi M. The significance of regular chest computed tomography in postoperative surveillance for surgically resected non-small cell lung cancer based on TNM 8th staging system. Gen Thorac Cardiovasc Surg 2024; 72:346-354. [PMID: 38143254 DOI: 10.1007/s11748-023-01991-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/05/2023] [Indexed: 12/26/2023]
Abstract
OBJECTIVES Although several societies recommend regular chest computed tomography (CT) scans for the surveillance of surgically resected non-small cell lung cancer (NSCLC), there is paucity of evidence to support these statements. This study aimed to clarify whether regular CT scans improved the prognosis of patients with surgically resected NSCLC based on TNM 8th classification. METHODS Patients with pathologic Stage 0-III NSCLC who underwent complete surgical resection other than sublobar resection procedures were enrolled in the study. For these patients, clinicopathological data and postoperative surveillance data were collected by the retrospective review of medical records. Patients were categorized into the chest X-ray (CXR) group or the CT group according to whether they were followed-up with basic examinations including CXR or basic examinations plus regular chest CT. Postoperative overall survival was compared between the two groups. RESULTS Six hundred sixty five patients were categorized into the CXR (n = 245) and CT (n = 420) groups. The clinicopathological backgrounds did not differ to a statistically significant extent. Recurrence was seen in 68 (27.3%) patients in the CXR group and 117 (27.8%) patients in the CT group. The 5-year overall survival rates of the two groups did not differ to a statistically significant extent (CXR, 76.5%; CT, 78.3%, P = 0.22). CONCLUSION Regular chest CT scans may not improve the prognosis of surgically resected NSCLC. Further study is warranted to precisely evaluate the benefit of CT-based postoperative surveillance of NSCLC.
Collapse
Affiliation(s)
- Jun Suzuki
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomohiro Miyoshi
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Kenta Tane
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Ken Onodera
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yutaro Koike
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Sakai
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Division of Chest Surgery, Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Joji Samejima
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Keiju Aokage
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| |
Collapse
|
42
|
Wang Y, Song Y, Wang R, Wu Y, Li M, Xu K, He R, Wang Z, Li Q, Kong FM(S, Wang T. Clinical factors and major pathological response after neoadjuvant chemoimmunotherapy in potentially resectable lung squamous cell carcinoma. Front Oncol 2024; 14:1265228. [PMID: 38680859 PMCID: PMC11045983 DOI: 10.3389/fonc.2024.1265228] [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: 07/22/2023] [Accepted: 03/18/2024] [Indexed: 05/01/2024] Open
Abstract
Objective Major pathological response (MPR) helps evaluate the prognosis of patients with lung squamous cell carcinoma (LUSC). However, the clinical factors that affect the achievement of MPR after neoadjuvant chemoimmunotherapy (NCIO) in patients with LUSC remain unclear. This study aimed to explore the clinical factors affecting the MPR after NCIO in patients with potentially resectable LUSC. Methods This retrospective study included patients with stage IIB-IIIC LUSC who underwent surgical resection after receiving NCIO at a center between March 2020 and November 2022. In addition to the postoperative pathological remission rate, sex, age, body mass index (BMI), smoking history, TNM stage, hematological and imaging test results, and other indicators were examined before NCIO. According to the pathological response rate of the surgically removed tumor tissue, the patients were split into MPR and non-MPR groups. Results In total, 91 LUSC patients who met the study's eligibility criteria were enrolled: 32 (35%) patients in the non-MPR group and 59 (65%) in the MPR group, which included 43 cases of pathological complete remission (pCR). Pre-treatment lymphocyte level (LY) (odds ratio [OR] =5.997), tumor burden (OR=0.958), N classification (OR=15.915), radiographic response (OR=11.590), pulmonary atelectasis (OR=5.413), and PD-L1 expression (OR=1.028) were independently associated with MPR (all P < 0.05). Based on these six independent predictors, we developed a nomogram model of prediction having an area under the curve (AUC) of 0.914 that is simple to apply clinically to predict the MPR. The MPR group showed greater disease-free survival (DFS) than the non-MPR group, according to the survival analysis (P < 0.001). Conclusion The MPR rate of NCIO for potentially resectable LUSC was 65%. LY, tumor burden, N classification, radiographic response, pulmonary atelectasis, and PD-L1 expression in patients with LUSC before NCIO were the independent and ideal predictors of MPR. The developed nomogram demonstrated a good degree of accuracy and resilience in predicting the MPR following NCIO, indicating that it is a useful tool for assuring customized therapy for patients with possibly resectable LUSC.
Collapse
Affiliation(s)
- Ye Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
- School of Graduate, Dalian Medical University, Dalian, China
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Runze Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Yu Wu
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
- School of Graduate, Dalian Medical University, Dalian, China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Ke Xu
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Rong He
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Zheng Wang
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Qingqing Li
- Department of Endoscopy, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Feng-Ming (Spring) Kong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
- Faculty of Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
43
|
Su T, Trang N, Zhu J, Kong L, Cheung D, Chou V, Ellis L, Huang C, Camden N, McHugh CA. GRAS1 non-coding RNA protects against DNA damage and cell death by binding and stabilizing NKAP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.20.545783. [PMID: 38645172 PMCID: PMC11030241 DOI: 10.1101/2023.06.20.545783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Non-coding RNA (ncRNA) gene products are involved in diverse biological processes including splicing, epigenetic regulation, gene expression, proliferation, and metabolism. The biological mechanisms by which ncRNAs contribute to cell survival remain poorly understood. We found that the Growth Regulator Antisense 1 (GRAS1) long non-coding RNA (lncRNA) transcript promotes growth in multiple human cell types by protecting against DNA damage. Knockdown of GRAS1 induced DNA damage and cell death, along with significant expression changes in DNA damage response, intrinsic apoptotic signaling, and cellular response to environmental stimulus genes. Extensive DNA damage occurred after GRAS1 knockdown, with numerous double strand breaks occurring in each cell. The number of cells undergoing apoptosis and with fragmented nuclei increased significantly after GRAS1 knockdown. We used RNA antisense purification and mass spectrometry (RAP-MS) to identify the NF-κB activating protein (NKAP) as a direct protein interaction partner of GRAS1 lncRNA. NKAP protein was degraded after GRAS1 knockdown, in a proteasome-dependent manner. Overexpression of GRAS1 or NKAP mitigated the DNA damage effects of GRAS1 knockdown. In summary, GRAS1 and NKAP directly interact to protect against DNA damage and cell death in multiple human cell lines.
Collapse
Affiliation(s)
| | | | - Jonathan Zhu
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Lingbo Kong
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Darin Cheung
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Vita Chou
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Lauren Ellis
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Calvin Huang
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Nichelle Camden
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Colleen A. McHugh
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
44
|
Zhang X, Ma L, Xue M, Sun Y, Wang Z. Advances in lymphatic metastasis of non-small cell lung cancer. Cell Commun Signal 2024; 22:201. [PMID: 38566083 PMCID: PMC10986052 DOI: 10.1186/s12964-024-01574-1] [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: 12/23/2023] [Accepted: 03/16/2024] [Indexed: 04/04/2024] Open
Abstract
Lung cancer is a deeply malignant tumor with high incidence and mortality. Despite the rapid development of diagnosis and treatment technology, abundant patients with lung cancer are still inevitably faced with recurrence and metastasis, contributing to death. Lymphatic metastasis is the first step of distant metastasis and an important prognostic indicator of non-small cell lung cancer. Tumor-induced lymphangiogenesis is involved in the construction of the tumor microenvironment, except promoting malignant proliferation and metastasis of tumor cells, it also plays a crucial role in individual response to treatment, especially immunotherapy. Thus, this article reviews the current research status of lymphatic metastasis in non-small cell lung cancer, in order to provide some insights for the basic research and clinical and translational application in this field.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Li Ma
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Man Xue
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yanning Sun
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Zhaoxia Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
45
|
Tian X, Liu X, Wang K, Wang R, Li Y, Qian K, Wang T, Zhao X, Liu L, Zhang PL, Xiong Y, Rui J, Chen R, Zhang Y. Postoperative ctDNA in indicating the recurrence risk and monitoring the effect of adjuvant therapy in surgical non-small cell lung cancer. Thorac Cancer 2024; 15:797-807. [PMID: 38409945 PMCID: PMC10995713 DOI: 10.1111/1759-7714.15251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a potential novel biomarker to predict molecular residual disease (MRD) in lung cancer after definitive treatment. Herein, we investigated the value of ctDNA in prognosing risk of relapse and monitoring the effect of adjuvant therapy in surgical non-small cell lung cancer (NSCLC). METHODS We enrolled 58 NSCLC patients in a real-world setting, and 58 tumor tissues and 325 plasma samples were analyzed. Tumor tissues and plasma samples were subjected to targeted next-generation sequencing (NGS) of 1021 cancer-related and ultra-deep targeted NGS covering 338 genes, respectively. RESULTS ctDNA was detected in 31.0% of cases at the first postoperative time, which was associated with advanced tumor stage, T stage and KEAP1 or GRIN2A mutations in tissues. ctDNA positivity at landmark and longitudinal indicated the shorter disease-free survival. For patients with ctDNA positivity at the first postoperative time, regardless of adjuvant therapy, all patients who were persistently ctDNA positive during postoperative surveillance had disease recurrence. Among the patients who were ctDNA negative, only two patients (15.4%, 2/13) receiving adjuvant therapy relapsed, while one patient (50.0%, 1/2) without adjuvant therapy relapsed. For the first postoperative ctDNA negative patients, the recurrence rate of patients with adjuvant therapy was and higher than without adjuvant therapy (22.6% [7/31] vs. 11.1% [1/9]). The patients who became ctDNA positive may also benefit from intervention therapy. CONCLUSION Postoperative ctDNA is a prognostic marker, and ctDNA-detection may facilitate personalized adjuvant therapy, and applying adjuvant therapy to the patients with detectable ctDNA could bring clinical benefits for them.
Collapse
Affiliation(s)
- Xiaoru Tian
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Xingsheng Liu
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Kai Wang
- Medical CenterGeneplus‐BeijingBeijingChina
| | - Ruotian Wang
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Yuanbo Li
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Kun Qian
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Tengteng Wang
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Xin Zhao
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Lei Liu
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Pei Long Zhang
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| | | | - Jinqiu Rui
- Medical CenterGeneplus‐BeijingBeijingChina
| | | | - Yi Zhang
- Department of Thoracic SurgeryXuanwu Hospital Capital Medical UniversityBeijingChina
| |
Collapse
|
46
|
Huo G, Song Y, Liu W, Cao X, Chen P. Osimertinib in the treatment of resected EGFR-mutated non-small cell lung cancer: a cost-effectiveness analysis in the United States. Front Pharmacol 2024; 15:1300183. [PMID: 38606181 PMCID: PMC11007098 DOI: 10.3389/fphar.2024.1300183] [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: 09/23/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Background: In the double-blind phase III ADAURA randomized clinical trial, adjuvant osimertinib showed a substantial overall survival benefit in patients with stage IB to IIIA, EGFR-mutated, completely resected non-small cell lung cancer (NSCLC). We conduct a cost-effectiveness analysis comparing the use of adjuvant osimertinib to placebo in patients with stage IB to IIIA, EGFR-mutated, resected NSCLC. Methods: Based on the results obtained from the ADAURA trial, a Markov model with three-state was employed to simulate patients who were administered either osimertinib or placebo until disease recurrence or completion of the study period (3 years). Quality-adjusted life-years (QALYs), lifetime costs, and incremental cost-effectiveness ratio (ICER) were calculated with a willingness-to-pay (WTP) threshold of $150,000 per QALY. Both univariate and probabilistic sensitivity analyses were carried out to explore the robustness of the model. Results: Osimertinib produced additional 1.59 QALYs with additional costs of $492,710 compared to placebo, giving rise to ICERs of $309,962.66/QALY. The results of the univariate sensitivity analysis indicated that the utility of disease-free survival (DFS), cost of osimertinib, and discount rate had the greatest impact on the outcomes. Probabilistic sensitivity analysis showed that osimertinib exhibited a 0% chance of being considered cost-effective for patients using a WTP threshold $150,000/QALY. Conclusion: In our model, osimertinib was unlikely to be cost-effective compared to placebo for stage IB to IIIA, EGFR-mutated, completely resected NSCLC patients from the perspective of a U.S. payer at a WTP threshold of $150,000 per QALY.
Collapse
Affiliation(s)
- Gengwei Huo
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Ying Song
- Department of Pharmacy, Jining No. 1 People’s Hospital, Jining, Shandong, China
| | - Wenjie Liu
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xuchen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
47
|
Huang M, Liu B, Li X, Li N, Yang X, Wang Y, Zhang S, Lu F, Li S, Yan S, Wu N. Beneficial implications of adjuvant chemotherapy for stage IB lung adenocarcinoma exhibiting elevated SUVmax in FDG-PET/CT: a retrospective study from a single center. Front Oncol 2024; 14:1367200. [PMID: 38529383 PMCID: PMC10961360 DOI: 10.3389/fonc.2024.1367200] [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: 01/08/2024] [Accepted: 02/26/2024] [Indexed: 03/27/2024] Open
Abstract
Background Controversy surrounds the efficacy of adjuvant chemotherapy (ACT) in the treatment of stage I lung adenocarcinoma (LUAD). The objective of this study was to examine the impact of the maximum standardized uptake value (SUVmax) as measured by 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) on the efficacy of ACT in patients diagnosed with stage I LUAD. Methods We scrutinized the medical records of 928 consecutive patients who underwent complete surgical resection for pathological stage I LUAD at our institution. The ideal cut-off value for primary tumor SUVmax in terms of disease-free survival (DFS) and overall survival (OS) was determined using the X-tile software. The Kaplan-Meier method and Cox regression analysis were used for survival analysis. Results Based on the SUVmax algorithm, the ideal cutoff values were determined to be 4.9 for DFS and 5.0 for OS. We selected 5.0 as the threshold because OS is the more widely accepted predictive endpoint. In a multivariate Cox regression analysis, SUVmax ≥ 5.0, problematic IB stage, and sublobectomy were identified as independent risk factors for poor DFS and OS. It is noteworthy that patients who were administered ACT had significantly longer DFS and OS than what was observed in the subgroup of patients with pathological stage IB LUAD and SUVmax ≥ 5.0 (p < 0.035 and p ≤ 0.046, respectively). However, there was no observed survival advantage for patients in stages IA or IB who had an SUVmax < 5.0. Conclusion The preoperative SUVmax of tumors served as an indicator of the impact of ACT in the context of completely resected pathological stage I LUAD. Notably, patients within the Stage IB category exhibiting elevated SUVmax levels emerged as a subgroup experiencing substantial benefits from postoperative ACT.
Collapse
Affiliation(s)
- Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yaqi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shanyuan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fangliang Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shaolei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
- State Key Laboratory of Molecular Oncology, Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
48
|
Zhang L, Du J, Wu X. A novel neoadjuvant therapy for early-stage non-small cell lung cancer in a mouse model. J Thorac Dis 2024; 16:1108-1117. [PMID: 38505061 PMCID: PMC10944762 DOI: 10.21037/jtd-23-1555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/29/2023] [Indexed: 03/21/2024]
Abstract
Background Lung cancer is the common malignancy with high mortality rate in the world. Even with curative resection for early-stage lung cancer patients, the rate of postoperative recurrence and metastasis is still high. Neoadjuvant nivolumab combined with chemotherapy leads to improved pathological complete response rate and event-free survival in resectable non-small cell lung cancer (NSCLC) patients. However, the neoadjuvant therapy is not only accompanied by grade 3 or above adverse events which resulting in the potential missing out on the window for curative surgery for the patients, but also has low efficacy especially in patients with low programmed death ligand 1 (PD-L1) expression. Hence, it is particularly important to explore innovative ways to inhibit tumour recurrence and metastasis. Methods In the present study, we investigated whether neoadjuvant therapy with intralesional Rose Bengal (RB) elicited specific immune responses compared with control group, and then the lung cancer mouse model was used to evaluated the immunological mechanism. Results The secondary Lewis lung cancer cells (LLCs) tumour growth was significantly suppressed by RB intralesional injection into subcutaneous tumour; the formation rate of secondary tumours induced by the B16 melanoma cell injection was 100%. Intralesional RB neoadjuvant therapy before surgical resection exhibited effectively enhanced T central memory cells (Tcm) and T memory stem cells (Tscm) + naïve T cells (Tn) infiltration, elicited stronger cytotoxic T lymphocyte (CTL) responses against LLCs, and displayed markedly higher proportions of splenic lymphocytes that produce tumour necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) upon restimulation in a lung cancer mouse model. Conclusions Based on our preclinical data, neoadjuvant therapy with intralesional RB injection generated immune memory and prevented the recurrence and metastasis of tumour in a lung cancer mouse model, which provides a new strategy for neoadjuvant treatment of early-stage NSCLC.
Collapse
Affiliation(s)
- Lanlin Zhang
- Department of Medical Oncology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiangyuan Du
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xianghua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
49
|
Hu Z, Sui Q, Jin X, Shan G, Huang Y, Yi Y, Zeng D, Zhao M, Zhan C, Wang Q, Lin Z, Lu T, Chen Z. IL6-STAT3-C/EBPβ-IL6 positive feedback loop in tumor-associated macrophages promotes the EMT and metastasis of lung adenocarcinoma. J Exp Clin Cancer Res 2024; 43:63. [PMID: 38424624 PMCID: PMC10903044 DOI: 10.1186/s13046-024-02989-x] [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: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Lung cancer is one of the most common tumors in the world, and metastasis is one of the major causes of tumor-related death in lung cancer patients. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are frequently associated with tumor metastasis in human cancers. However, the regulatory mechanisms of TAMs in lung cancer metastasis remain unclear. METHODS Single-cell sequencing analysis of lung cancer and normal tissues from public databases and from 14 patients who underwent surgery at Zhongshan Hospital was performed. In vitro co-culture experiments were performed to evaluate the effects of TAMs on lung cancer migration and invasion. Changes in the expression of IL-6, STAT3, C/EBPΒ, and EMT pathway were verified using RT-qPCR, western blotting, and immunofluorescence. Dual luciferase reporter assays and ChIP were used to reveal potential regulatory sites on the transcription factor sets. In addition, the effects of TAMs on lung cancer progression and metastasis were confirmed by in vivo models. RESULTS TAM infiltration is associated with tumor progression and poor prognosis. IL-6 secreted by TAMs can activate the JAK2/STAT3 pathway through autocrine secretion, and STAT3 acts as a transcription factor to activate the expression of C/EBPβ, which further promotes the transcription and expression of IL-6, forming positive feedback loops for IL6-STAT3-C/EBPβ-IL6 in TAMs. IL-6 secreted by TAMs promotes lung cancer progression and metastasis in vivo and in vitro by activating the EMT pathway, which can be attenuated by the use of JAK2/STAT3 pathway inhibitors or IL-6 monoclonal antibodies. CONCLUSIONS Our data suggest that TAMs promote IL-6 expression by forming an IL6-STAT3-C/EBPβ-IL6 positive feedback loop. Released IL-6 can induce the EMT pathway in lung cancer to enhance migration, invasion, and metastasis. The use of IL-6-neutralizing antibody can partially counteract the promotion of LUAD by TAMs. A novel mechanism of macrophage-promoted tumor progression was revealed, and the IL6-STAT3-C/EBPβ-IL6 signaling cascade may be a potential therapeutic target against lung cancer.
Collapse
Affiliation(s)
- Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Xing Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Tao Lu
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital / Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences / Cancer Hospital Affiliated to Shanxi Medical University, No. 3 Gongren Xin Jie, Xinghualing District, Taiyuan, 030013, Shanxi Province, China.
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
50
|
Thamrongjirapat T, Muntham D, Incharoen P, Trachu N, Sae-Lim P, Sarachai N, Khiewngam K, Monnamo N, Kantathut N, Ngodngamthaweesuk M, Ativitavas T, Chansriwong P, Nitiwarangkul C, Ruangkanchanasetr R, Kositwattanarerk A, Sirachainan E, Dejthevaporn T, Reungwetwattana T. Molecular alterations and clinical prognostic factors in resectable non-small cell lung cancer. BMC Cancer 2024; 24:200. [PMID: 38347487 PMCID: PMC10863204 DOI: 10.1186/s12885-024-11934-2] [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: 08/28/2023] [Accepted: 01/29/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND EGFR inhibitor and immunotherapy have been approved for adjuvant treatment in resectable non-small cell lung cancer (NSCLC). Limited reports of molecular and clinical characteristics as prognostic factors in NSCLC have been published. METHODS Medical records of patients with resectable NSCLC stage I-III diagnosed during 2015-2020 were reviewed. Real time-PCR (RT-PCR) was performed for EGFR mutations (EGFRm). Immunohistochemistry staining was conducted for ALK and PD-L1 expression. Categorical variables were compared using chi-square test and Fisher's exact test. Survival analysis was done by cox-regression method. RESULTS Total 441 patients were included. The prevalence of EGFRm, ALK fusion, and PD-L1 expression were 57.8%, 1.9%, and 20.5% (SP263), respectively. The most common EGFRm were Del19 (43%) and L858R (41%). There was no significant difference of recurrence free survival (RFS) by EGFRm status whereas patients with PD-L1 expression (PD-L1 positive patients) had lower RFS compared to without PD-L1 expression (PD-L1 negative patients) (HR = 1.75, P = 0.036). Patients with both EGFRm and PD-L1 expression had worse RFS compared with EGFRm and PD-L1 negative patients (HR = 3.38, P = 0.001). Multivariable analysis showed higher CEA at cut-off 3.8 ng/ml, pT4, pN2, pStage II, and margin were significant poor prognostic factors for RFS in the overall population, which was similar to EGFRm population (exception of pT and pStage). Only pStage was a significant poor prognostic factor for PD-L1 positive patients. The predictive score for predicting of recurrence were 6 for all population (63% sensitivity and 86% specificity) and 5 for EGFRm population (62% sensitivity and 93% specificity). CONCLUSION The prevalence and types of EGFRm were similar between early stage and advanced stage NSCLC. While lower prevalence of PD-L1 expression was found in early stage disease. Patients with both EGFRm and PD-L1 expression had poorer outcome. Thus PD-L1 expression would be one of the prognostic factor in EGFRm patients. Validation of the predictive score should be performed in a larger cohort.
Collapse
Affiliation(s)
- T Thamrongjirapat
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - D Muntham
- Department of Mathematics, Faculty of Science and Technology, Rajamangala University of Technology Suvarnabhumi, Bangkok, Thailand
| | - P Incharoen
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - N Trachu
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - P Sae-Lim
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - N Sarachai
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - K Khiewngam
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - N Monnamo
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - N Kantathut
- Division of Thoracic Surgery, Department of Surgery, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - M Ngodngamthaweesuk
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Division of Thoracic Surgery, Department of Surgery, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - T Ativitavas
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - P Chansriwong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - C Nitiwarangkul
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Division of Diagnostic Radiology, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - R Ruangkanchanasetr
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Radiation and Oncology Unit, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - A Kositwattanarerk
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Division of Nuclear Medicine, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - E Sirachainan
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - T Dejthevaporn
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - T Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
- Ramathibodi Lung Cancer Consortium (RLC), Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|