1
|
Song Y, Jiang YX, Guan JY, Jiang JB, Xu MS, Zhong XY, He LN, Ren ZY, Liao Y, Liu F, Jiang YJ, Hu S, Guo W, Zhao TX, Liu XY, Gu JY, Shi YF, Luo HH, Wang K, Xiao JY. Fangchinoline-mediated autophagy inhibition amplifies antigen presentation and PD-1 blockade efficacy in lung cancer. Acta Pharmacol Sin 2025:10.1038/s41401-025-01541-7. [PMID: 40269245 DOI: 10.1038/s41401-025-01541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025]
Abstract
Cancer cells frequently exhibit MHC-I deficiency, impairing immune-mediated cytotoxicity even in the presence of PD-1 checkpoint inhibition. To date, no clinically approved therapies exist that can upregulate MHC-I expression to boost immune responses against cancer cells. Emerging evidence has shown that autophagy plays a role in MHC-I molecule degradation, contributing to reduced recognition of cancer cells by CD8+ T cells. We previously report that fangchinoline, a bisbenzylisoquinoline alkaloid derived from Chinese herb, is a novel autophagy inhibitor with an adjuvant of chemotherapy against lung cancer. In this study we investigated the modulatory effects of PD-1 blockade combined with fangchinoline on CD8+ T cells within the tumor microenvironment of lung cancer. We showed an inverse correlation between elevated autophagic activity and decreased MHC-I surface expression-a phenomenon often associated with poor clinical efficacies-in various human lung cancer cell lines (NCI-H1299, NCI-H1975, A549, NCI-H1650 and NCI-H446) compared with normal bronchial epithelial cells lung cancer. Knockdown of ATG4 and ATG5 resulted in increased MHC-I expression and enhanced tumor antigen presentation in NCI-H1975, NCI-H1299 and A549 cells. As autophagy receptors were crucial for transporting proteins to autophagosomes for degradation, we sequentially silenced various autophagy receptors and found that NDP52 knockdown specifically restored MHC-I expression, suggesting that NDP52-mediated autophagy might contribute to MHC-I degradation, and autophagy inhibition might enhance immune-mediated cancer cell death. We showed that pretreatment of LLC-OVA cells with the autophagy inhibitor fangchinoline (1.25, 2.5, 5 μM) followed by coculture with CD8+ T cells, dose-dependently enhanced immune killing. In both in vitro and in vivo experiments, we showed that fangchinoline combined with anti-PD-1 therapy significantly increased CD8+ T cell-mediated cytotoxicity. In conclusion, this study highlights NDP52 as a key autophagy receptor involved in MHC-I degradation and provides a new insight into tumor immune evasion. Combining autophagy inhibition with immunotherapy may be a promising therapeutic strategy for anticancer immunity enhancement.
Collapse
Affiliation(s)
- Yue Song
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yu-Xiao Jiang
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jie-Ying Guan
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jun-Bo Jiang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Man-Si Xu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xue-Ying Zhong
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Li-Na He
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhen-Yang Ren
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuan Liao
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Fang Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yan-Jun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Shan Hu
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wei Guo
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ting-Xiu Zhao
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiao-Yi Liu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jiang-Yong Gu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ya-Fei Shi
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huan-Huan Luo
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Kun Wang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jian-Yong Xiao
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Siemiątkowska A, Kuźnar-Kamińska B, Bryl M, Kosicka-Noworzyń K, Przybył P, Gołda-Gocka I, Główka FK. Low serum neopterin early indicates durable benefits of atezolizumab and pembrolizumab therapy in advanced lung cancer. Sci Rep 2025; 15:13078. [PMID: 40240475 PMCID: PMC12003904 DOI: 10.1038/s41598-025-97792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer is a state of immunological imbalance associated with chronic inflammation and local immunosuppression. Introducing immune checkpoint inhibitors was a breakthrough in cancer treatment. However, the treatment outcomes remain unsatisfactory, and many patients still progress after the initial response. The study aimed to assess whether serum neopterin (NEO), an indicator of cellular immune activation, could be used as a predictor of the long-term benefits of drugs blocking the programmed cell death protein 1 pathway (anti-PD-1/PD-L1 drugs). We enrolled 103 patients with non-small cell lung cancer (NSCLC) treated with anti-PD-1/PD-L1s. Serum was collected at baseline and at the end of each treatment cycle for the first three months of immunotherapy. NEO concentrations were determined with a validated high-performance liquid chromatography assay and correlated with treatment outcomes. Low-NEO status (i.e., serum NEO levels ≤ 71.65 nM at the end of the 3rd treatment cycle and ≤ 66.84 nM at the end of the 4th treatment cycle) increased the odds of ≥ 12-month benefits (odds ratio, OR = 11.70, p < 0.001), and decreased the hazard of NSCLC progression (hazard ratio, HR = 0.327, p < 0.001) and treatment failure (adjusted HR = 0.450, p < 0.05). Patients with low-NEO status had three times longer progression-free survival (PFS, 17.3 vs. 5.9 months) and three times longer time to treatment failure (TTF, 16.3 vs. 5.5 months) compared to other patients. Baseline NEO levels could not discriminate between patients who had and lacked the long-term benefits of treatment. In conclusion, the on-treatment serum NEO concentrations could be a biomarker of the long-term benefits of the anti-PD-1/PD-L1 treatment in advanced NSCLC.
Collapse
MESH Headings
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/blood
- Lung Neoplasms/pathology
- Lung Neoplasms/mortality
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Male
- Middle Aged
- Aged
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/blood
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Neopterin/blood
- Immune Checkpoint Inhibitors/therapeutic use
- Biomarkers, Tumor/blood
- Treatment Outcome
- Adult
- Aged, 80 and over
Collapse
Affiliation(s)
- Anna Siemiątkowska
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznań, Poland.
| | - Barbara Kuźnar-Kamińska
- Department of Pulmonology, Allergology and Respiratory Oncology, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569, Poznań, Poland
| | - Maciej Bryl
- Department of Clinical Oncology with the Subdepartment of Diurnal Chemotherapy, Wielkopolska Center of Pulmonology and Thoracic Surgery, 62 Szamarzewskiego Street, 60-569, Poznań, Poland
| | - Katarzyna Kosicka-Noworzyń
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznań, Poland
| | - Paulina Przybył
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznań, Poland
| | - Iwona Gołda-Gocka
- Department of Clinical Oncology with the Subdepartment of Diurnal Chemotherapy, Wielkopolska Center of Pulmonology and Thoracic Surgery, 62 Szamarzewskiego Street, 60-569, Poznań, Poland
| | - Franciszek K Główka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznań, Poland
| |
Collapse
|
3
|
Lu J, Liu X, Ji X, Jiang Y, Zuo A, Guo Z, Yang S, Peng H, Sun F, Lu D. Predicting PD-L1 status in NSCLC patients using deep learning radiomics based on CT images. Sci Rep 2025; 15:12495. [PMID: 40216830 PMCID: PMC11992188 DOI: 10.1038/s41598-025-91575-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/21/2025] [Indexed: 04/14/2025] Open
Abstract
Radiomics refers to the utilization of automated or semi-automated techniques to extract and analyze numerous quantitative features from medical images, such as computerized tomography (CT) or magnetic resonance imaging (MRI) scans. This study aims to develop a deep learning radiomics (DLR)-based approach for predicting programmed death-ligand 1 (PD-L1) expression in patients with non-small cell lung cancer (NSCLC). Data from 352 NSCLC patients with known PD-L1 expression were collected, of which 48.29% (170/352) were tested positive for PD-L1 expression. Tumor regions of interest (ROI) were semi-automatically segmented based on CT images, and DL features were extracted using Residual Network 50. The least absolute shrinkage and selection operator (LASSO) algorithm was used for feature selection and dimensionality reduction. Seven algorithms were used to build models, and the most optimal ones were identified. A combined model integrating DLR with clinical data was also developed. The predictive performance of each model was evaluated using the area under the curve (AUC) of the receiver operating characteristic (ROC) curve analysis. The DLR model, based on CT images, demonstrated an AUC of 0.85 (95% confidence interval (CI), 0.82-0.88), sensitivity of 0.80 (0.74-0.85), and specificity of 0.73 (0.70-0.77) for predicting PD-L1 status. The integrated model exhibited superior performance, with an AUC of 0.91 (0.87-0.95), sensitivity of 0.85 (0.82-0.89), and specificity of 0.75 (0.72-0.80). Our findings indicate that the DLR model holds promise as a valuable tool for predicting the PD-L1 status in patients with NSCLC, which can greatly assist in clinical decision-making and the selection of personalized treatment strategies.
Collapse
Affiliation(s)
- Jiameng Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
- Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau Special Administrative Region, People's Republic of China
| | - Xinyi Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Xiaoqing Ji
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Yunxiu Jiang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Anli Zuo
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Zihan Guo
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Shuran Yang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Haiying Peng
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Yibin City, 644002, Yibin, People's Republic of China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, Jining No.1 People's Hospital, 272000, Jining, People's Republic of China
| | - Degan Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
4
|
Su H, Shen J, Gao C, Zhao Y, Deng W, Qin B, Zhang X, Lai J, Wang Q, Dou J, Guo M. Epsin3 promotes non-small cell lung cancer progression via modulating EGFR stability. Cell Biosci 2025; 15:14. [PMID: 39910656 PMCID: PMC11800460 DOI: 10.1186/s13578-025-01358-1] [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: 10/11/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND The abnormal expression and overactivation of the epidermal growth factor receptor (EGFR), a typical cancer marker for non-small cell lung cancer (NSCLC), are closely related to the tumorigenesis and progression of NSCLC. However, the endocytosis mechanism of EGFR in lung cancer is not yet known. Epsin3 (EPN3), a member of the endocytic adaptor protein family, is essential for the endocytosis of multiple receptors. In this study, we aimed to investigate the role of EPN3 in modulating EGFR function, its effects on NSCLC progression, and its potential involvement in tyrosine kinase inhibitor (TKI) resistance, which remains a significant hurdle in NSCLC treatment. RESULTS Our findings revealed that the expression of EPN3 is significantly up-regulated in NSCLC patients. Elevated EPN3 expression was proportional to shorter overall survival in patients with NSCLC. Functional analyses revealed that EPN3 directly interacts with EGFR, enhancing its recycling to the plasma membrane and preventing its degradation via the lysosomal pathway. This stabilization of EGFR led to sustained downstream signalling, promoting NSCLC cell proliferation and migration. Notably, mutations in the EGFR tyrosine kinase domain, which typically confer resistance to TKIs, did not alter the regulatory effect of EPN3. CONCLUSIONS EPN3 enhances EGFR signalling by promoting its recycling and stability, contributing to NSCLC progression and TKI resistance. Targeting EPN3 could offer a novel therapeutic strategy to overcome drug resistance in EGFR-driven NSCLC.
Collapse
Affiliation(s)
- Huiling Su
- State Key Laboratory of Natural Medicines, School of Life Science & Technology, Pharmaceutical University, 210009, Nanjing, China
| | - Jie Shen
- State Key Laboratory of Natural Medicines, School of Life Science & Technology, Pharmaceutical University, 210009, Nanjing, China
| | - Chenzi Gao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, 210029, Nanjing, China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, School of Life Science & Technology, Pharmaceutical University, 210009, Nanjing, China
| | - Wanyu Deng
- College of Life Science, Shangrao Normal University, 334001, Shangrao, China
| | - Bo Qin
- Shaoxing Women and Children's Hospital, 312000, Shaoxing, China
| | - Xin Zhang
- GeneMind Biosciences Company Limited, 518001, Shenzhen, China
| | - Juan Lai
- GeneMind Biosciences Company Limited, 518001, Shenzhen, China
| | - Qian Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, 210029, Nanjing, China.
| | - Jie Dou
- State Key Laboratory of Natural Medicines, School of Life Science & Technology, Pharmaceutical University, 210009, Nanjing, China.
| | - Min Guo
- State Key Laboratory of Natural Medicines, School of Life Science & Technology, Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
5
|
Jiang L, Jiang S, Miao W, Shen Y, Bolotina L, Zhu H, Zou N, Tian Y, Pan H, Huang J, Ryabov A, Luo Q. Clinical Characteristics and Management of Checkpoint Inhibitor Pneumonitis in Non-Small-Cell Lung Cancer Patients After Neoadjuvant Immunotherapy. Clin Lung Cancer 2025; 26:e91-e98. [PMID: 39578169 DOI: 10.1016/j.cllc.2024.10.012] [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: 05/13/2024] [Revised: 09/01/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE Neoadjuvant immunotherapy with checkpoint inhibitors has shown promising results for non-small-cell lung cancer (NSCLC) patients. However, it has been associated with immune-related adverse events (irAEs), including checkpoint inhibitor pneumonitis (CIP), which can be life-threatening. METHODS This retrospective study analysed the medical records of 197 NSCLC patients who underwent neoadjuvant checkpoint inhibitor therapy to investigate the incidence, clinical characteristics, and management of CIP. RESULTS Of the 197 patients, 24 (12.2%) developed CIP. The majority of patients presented with respiratory symptoms, and ground-glass opacities were the most common radiographic finding. Patients with CIP had a longer duration of immunotherapy and a higher baseline C-reactive protein level than those without CIP. Most cases were mild to moderate in severity (Grade 1: 11, Grade 2: 6, Grade 3: 5) and managed with corticosteroids, while 2 patients of Grade 4 developed severe respiratory failure requiring mechanical ventilation. No patient died due to respiratory failure. CONCLUSIONS CIP was identified as a potential complication of neoadjuvant treatment with checkpoint inhibitors in patients with resectable NSCLC. Therefore, close monitoring for CIP and prompt recognition and management of symptoms are essential for the safe use of checkpoint inhibitors in NSCLC patients. While the study also found that such neoadjuvant treatment can induce a major pathological response in a significant proportion of these patients, further research is required to fully understand and manage the risk factors of CIP in this patient population.
Collapse
Affiliation(s)
- Long Jiang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shanshan Jiang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China; State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Shanghai, China
| | - Wang Miao
- Department of Thoracic Surgery, The Third People's Hospital of Zhengzhou, Zhengzhou, China
| | - Yaofeng Shen
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Larisa Bolotina
- Thoracoabdominal department оf Moscow Oncology Research Institute named after P. Hertsen-branch of Federal State Budgetary Institution "National Medical Research Radiological Centre" of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Hongda Zhu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningyuan Zou
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Tian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanbo Pan
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Huang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Andrey Ryabov
- Thoracoabdominal department оf Moscow Oncology Research Institute named after P. Hertsen-branch of Federal State Budgetary Institution "National Medical Research Radiological Centre" of the Ministry of Health of the Russian Federation, Moscow, Russia.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Yazdan M, Naghib SM. Smart Ultrasound-responsive Polymers for Drug Delivery: An Overview on Advanced Stimuli-sensitive Materials and Techniques. Curr Drug Deliv 2025; 22:283-309. [PMID: 38288800 DOI: 10.2174/0115672018283792240115053302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 04/11/2025]
Abstract
In recent years, a notable advancement has occurred in the domain of drug delivery systems via the integration of intelligent polymers that respond to ultrasound. The implementation of this groundbreaking methodology has significantly revolutionised the controlled and precise delivery of therapeutic interventions. An in-depth investigation is conducted into the most recent developments in ultrasonic stimulus-responsive materials and techniques for the purpose of accomplishing precise medication administration. The investigation begins with an exhaustive synopsis of the foundational principles underlying drug delivery systems that react to ultrasonic stimuli, focusing specifically on the complex interplay between polymers and ultrasound waves. Significant attention is devoted to the development of polymers that demonstrate tailored responsiveness to ultrasound, thereby exemplifying their versatility in generating controlled drug release patterns. Numerous classifications of intelligent polymers are examined in the discussion, including those that react to variations in temperature, pH, and enzymes. When coupled with ultrasonic stimuli, these polymers offer a sophisticated framework for the precise manipulation of drug release in terms of both temporal and spatial dimensions. The present study aims to examine the synergistic effects of responsive polymers and ultrasound in overcoming biological barriers such as the blood-brain barrier and the gastrointestinal tract. By doing so, it seeks to shed light on the potential applications of these materials in intricate clinical scenarios. The issues and future prospects of intelligent ultrasound-responsive polymers in the context of drug delivery are critically analysed in this article. The objective of this study is to offer valuable perspectives on the challenges that must be overcome to enable the effective implementation of these technologies. The primary objective of this comprehensive review is to furnish researchers, clinicians, and pharmaceutical scientists with a wealth of information that will serve as a guide for forthcoming developments in the development and enhancement of intelligent drug delivery systems that employ ultrasound-responsive polymers to attain superior therapeutic outcomes.
Collapse
Affiliation(s)
- Mostafa Yazdan
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| |
Collapse
|
7
|
Fang R, Chen Y, Huang B, Wang Z, Zhu X, Liu D, Sun W, Chen L, Zhang M, Lyu K, Lei W. Predicting response to PD-1 inhibitors in head and neck squamous cell carcinomas using peripheral blood inflammatory markers. Transl Oncol 2025; 51:102222. [PMID: 39616985 DOI: 10.1016/j.tranon.2024.102222] [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: 07/14/2024] [Revised: 11/15/2024] [Accepted: 11/23/2024] [Indexed: 12/11/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) treatment has the potential to induce durable disease remission. However, the current combined positive score (CPS) is insufficient accurate for predicting which patients will benefit from it. In the present study, a real-world retrospective study was conducted on 56 patients of HNSCC who received ICI treatment. Then the treatment that patient received and levels of pre-treatment blood inflammatory markers (NLR, MLR and PLR) were identified to develop a model for predicting immunotherapy response. Notably, the model achieved an area under the curve (AUC) of 0.877 (95 % CI 0.769-0.985) , providing a larger net benefit than the CPS marker (AUC=0.614, 95 % CI 0.466-0.762). Furthermore, the internal validation of the prediction model showed a C-index of 0.835. Patients with high score of the model would get improved PFS than those with low score. Therefore, the prediction model for patients with local advanced or R/M HNSCC receiving ICI treatment, which represented an better efficient prediction of immunotherapy response than CPS marker.
Collapse
Affiliation(s)
- Ruihua Fang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Yi Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Bixue Huang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Zhangfeng Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Xiaolin Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Dawei Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Wei Sun
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Lin Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Minjuan Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China
| | - Kexing Lyu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China.
| | - Wenbin Lei
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, PR China.
| |
Collapse
|
8
|
Janzen I, Ho C, Melosky B, Ye Q, Li J, Wang G, Lam S, MacAulay C, Yuan R. Machine Learning and Computed Tomography Radiomics to Predict Disease Progression to Upfront Pembrolizumab Monotherapy in Advanced Non-Small-Cell Lung Cancer: A Pilot Study. Cancers (Basel) 2024; 17:58. [PMID: 39796687 PMCID: PMC11719007 DOI: 10.3390/cancers17010058] [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: 11/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pembrolizumab monotherapy is approved in Canada for first-line treatment of advanced NSCLC with PD-L1 ≥ 50% and no EGFR/ALK aberrations. However, approximately 55% of these patients do not respond to pembrolizumab, underscoring the need for the early intervention of non-responders to optimize treatment strategies. Distinguishing the 55% sub-cohort prior to treatment is a real-world dilemma. METHODS In this retrospective study, we analyzed two patient cohorts treated with pembrolizumab monotherapy (training set: n = 97; test set: n = 17). The treatment response was assessed using baseline and follow-up CT scans via RECIST 1.1 criteria. RESULTS A logistic regression model, incorporating pre-treatment CT radiomic features of lung tumors and clinical variables, achieved high predictive accuracy (AUC: 0.85 in training; 0.81 in testing, 95% CI: 0.63-0.99). Notably, radiomic features from the peritumoral region were found to be independent predictors, complementing the standard CT evaluations and other clinical characteristics. CONCLUSIONS This pragmatic model offers a valuable tool to guide first-line treatment decisions in NSCLC patients with high PD-L1 expression and has the potential to advance personalized oncology and improve timely disease management.
Collapse
Affiliation(s)
- Ian Janzen
- Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z Il3, Canada; (I.J.)
- Interdisciplinary Oncology Program, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Cheryl Ho
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Medical Oncology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Barbara Melosky
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Medical Oncology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Qian Ye
- Department of Statistics, Faculty of Science, University of British Columbia, 2329 West Mall, Vancouver, BC V6T 1Z4, Canada
| | - Jessica Li
- Department of Radiology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Gang Wang
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Stephen Lam
- Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z Il3, Canada; (I.J.)
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Respirology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Calum MacAulay
- Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z Il3, Canada; (I.J.)
- Department of Pathology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Ren Yuan
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Radiology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| |
Collapse
|
9
|
Watanabe SI, Yotsukura M, Miyoshi T, Hattori A, Isaka T, Maniwa T, Isaka M, Yoshioka H, Endo M, Mimae T, Tsutani Y, Nakagawa K, Aokage K. Updated review of perioperative treatment for non-small-cell lung cancer in the new era of immune checkpoint inhibitors: past, present, and future. Jpn J Clin Oncol 2024; 54:1244-1253. [PMID: 39163130 DOI: 10.1093/jjco/hyae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
The perioperative treatments for non-small cell lung cancer (NSCLC) should control both local and microscopic systemic disease, because the survival of patients with NSCLC who underwent surgical resection alone has been dismal except in stage IA patients. One way to improve surgical outcome is the administration of chemotherapy before or after the surgical procedure. During the last two decades, many clinical studies have focused on developing optimal adjuvant or neoadjuvant cisplatin-based chemotherapy regimens that can be combined with surgical treatment and/or radiotherapy. Based on the results of those clinical studies, multimodality therapy has been considered to be an appropriate treatment approach for locally advanced NSCLC patients. When nodal involvement is discovered postoperatively, adjuvant cisplatin-based chemotherapy has conferred an overall survival benefit. More recently, neoadjuvant and/or adjuvant use of immunotherapy adding to the cisplatin-based chemotherapy has been revealed to improve survival of the patients with locally advanced NSCLC in many large-scale clinical trials; although, optimal treatment strategies are still evolving.
Collapse
Affiliation(s)
- Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Masaya Yotsukura
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Tomohiro Miyoshi
- Division of Thoracic Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Aritoshi Hattori
- Division of General Thoracic Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Tetsuya Isaka
- Department of Thoracic Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Tomohiro Maniwa
- Department of Thoracic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Mitsuhiro Isaka
- Department of Thoracic Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University Hospital, Osaka, Japan
| | - Makoto Endo
- Department of Thoracic Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Takahiro Mimae
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Yasuhiro Tsutani
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuo Nakagawa
- Department of Thoracic Surgery, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keiju Aokage
- Division of Thoracic Surgery, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
10
|
Fan J, Zhang T. Multi-index comprehensive evaluation of the efficacy and response mechanism of immunotherapy in non-small cell lung cancer. Clin Transl Oncol 2024; 26:3124-3130. [PMID: 38822977 DOI: 10.1007/s12094-024-03519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/12/2024] [Indexed: 06/03/2024]
Abstract
OBJECTIVE This research conducted multi-index comprehensive evaluations of the immunotherapeutic efficacy and response in non-small cell lung cancer (NSCLC). METHODS Forty-five patients with epidermal growth factor receptor (EGFR)/anaplastic lymphoma kinase (ALK) wild-type advanced NSCLC who received immunotherapy were included. Immunohistochemistry was adopted to detect the expression levels of programmed death ligand 1 (PD-L1) with X-ray cross-complementing protein 1 (XRCC1) and excision repair cross-complementing group 1 (ERCC1) proteins in tumor tissues. Flow cytometry was utilized to measure the levels of T-cell subsets in peripheral blood before and after treatment. PCR-RELP method was employed to evaluate XRCC1 and ERCC1 gene polymorphisms in peripheral blood. According to the treatment effect, patients evaluated as complete response (CR), partial response (PR), and stable disease (SD) were categorized into the immune response group, and patients evaluated as progressive disease (PD) were categorized into the immune unresponsive group. The correlation between PD-L1 protein expression, XRCC1 and ERCC1 protein expression, gene polymorphisms, T-cell subpopulation levels, and treatment efficacy was analyzed. RESULTS The therapeutic efficacy of patients with positive PD-L1 expression was better than that of patients with negative PD-L1 expression (P < 0.05). After treatment, peripheral blood CD3+ and CD4+ cell levels and Thl/Th2 cell levels were higher and CD8+ T cells were lower in the immune response group than in the immune unresponsive group (P < 0.05). Among the patients in the immune response group, peripheral blood CD3+ and CD4+ cell levels were higher and CD8+ T cells were lower in patients with positive PD-L1 expression than in patients with negative PD-L1 expression (P < 0.05). In the XRCC1 gene, the proportion of patients in the immune response group carrying the Arg/Trp + Trp/Trp genotype was higher than that of patients in the immune unresponsive group (P < 0.05). In the ERCC1 gene, the proportion of patients in the immune response group carrying the C/T + T/T genotype was higher than that of patients in the immune unresponsive group (P < 0.05). The positive expression rates of XRCC1 and ERCC1 in patients in the immune unresponsive group were higher than those in the immune response group (P < 0.05). CONCLUSION PD-L1 protein expression, XRCC1 and ERCC1 protein expression, and gene polymorphisms are associated with immunotherapy outcome in EGFR/ALK wild-type advanced NSCLC patients, and may be biological indicators for predicting immunotherapy outcome in EGFR/ALK wild-type advanced NSCLC patients.
Collapse
Affiliation(s)
- Jieqiong Fan
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
- Department of Oncology, The Chongqing University Three Gorges Hospital, The Chongqing Three Gorges Central Hospital, 165 Xincheng Road, Wanzhou District, Chongqing, 404000, China
| | - Tao Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
11
|
Reina C, Šabanović B, Lazzari C, Gregorc V, Heeschen C. Unlocking the future of cancer diagnosis - promises and challenges of ctDNA-based liquid biopsies in non-small cell lung cancer. Transl Res 2024; 272:41-53. [PMID: 38838851 DOI: 10.1016/j.trsl.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
The advent of liquid biopsies has brought significant changes to the diagnosis and monitoring of non-small cell lung cancer (NSCLC), presenting both promise and challenges. Molecularly targeted drugs, capable of enhancing survival rates, are now available to around a quarter of NSCLC patients. However, to ensure their effectiveness, precision diagnosis is essential. Circulating tumor DNA (ctDNA) analysis as the most advanced liquid biopsy modality to date offers a non-invasive method for tracking genomic changes in NSCLC. The potential of ctDNA is particularly rooted in its ability to furnish comprehensive (epi-)genetic insights into the tumor, thereby aiding personalized treatment strategies. One of the key advantages of ctDNA-based liquid biopsies in NSCLC is their ability to capture tumor heterogeneity. This capability ensures a more precise depiction of the tumor's (epi-)genomic landscape compared to conventional tissue biopsies. Consequently, it facilitates the identification of (epi-)genetic alterations, enabling informed treatment decisions, disease progression monitoring, and early detection of resistance-causing mutations for timely therapeutic interventions. Here we review the current state-of-the-art in ctDNA-based liquid biopsy technologies for NSCLC, exploring their potential to revolutionize clinical practice. Key advancements in ctDNA detection methods, including PCR-based assays, next-generation sequencing (NGS), and digital PCR (dPCR), are discussed, along with their respective strengths and limitations. Additionally, the clinical utility of ctDNA analysis in guiding treatment decisions, monitoring treatment response, detecting minimal residual disease, and identifying emerging resistance mechanisms is examined. Liquid biopsy analysis bears the potential of transforming NSCLC management by enabling non-invasive monitoring of Minimal Residual Disease and providing early indicators for response to targeted treatments including immunotherapy. Furthermore, considerations regarding sample collection, processing, and data interpretation are highlighted as crucial factors influencing the reliability and reproducibility of ctDNA-based assays. Addressing these challenges will be essential for the widespread adoption of ctDNA-based liquid biopsies in routine clinical practice, ultimately paving the way toward personalized medicine and improved outcomes for patients with NSCLC.
Collapse
Affiliation(s)
- Chiara Reina
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Berina Šabanović
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Chiara Lazzari
- Department of Medical Oncology, Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Vanesa Gregorc
- Department of Medical Oncology, Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Christopher Heeschen
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy;.
| |
Collapse
|
12
|
Hoshi R, Gorospe KA, Labouta HI, Azad T, Lee WL, Thu KL. Alternative Strategies for Delivering Immunotherapeutics Targeting the PD-1/PD-L1 Immune Checkpoint in Cancer. Pharmaceutics 2024; 16:1181. [PMID: 39339217 PMCID: PMC11434872 DOI: 10.3390/pharmaceutics16091181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
The programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint constitutes an inhibitory pathway best known for its regulation of cluster of differentiation 8 (CD8)+ T cell-mediated immune responses. Engagement of PD-L1 with PD-1 expressed on CD8+ T cells activates downstream signaling pathways that culminate in T cell exhaustion and/or apoptosis. Physiologically, these immunosuppressive effects exist to prevent autoimmunity, but cancer cells exploit this pathway by overexpressing PD-L1 to facilitate immune escape. Intravenously (IV) administered immune checkpoint inhibitors (ICIs) that block the interaction between PD-1/PD-L1 have achieved great success in reversing T cell exhaustion and promoting tumor regression in various malignancies. However, these ICIs can cause immune-related adverse events (irAEs) due to off-tumor toxicities which limits their therapeutic potential. Therefore, considerable effort has been channeled into exploring alternative delivery strategies that enhance tumor-directed delivery of PD-1/PD-L1 ICIs and reduce irAEs. Here, we briefly describe PD-1/PD-L1-targeted cancer immunotherapy and associated irAEs. We then provide a detailed review of alternative delivery approaches, including locoregional (LDD)-, oncolytic virus (OV)-, nanoparticle (NP)-, and ultrasound and microbubble (USMB)-mediated delivery that are currently under investigation for enhancing tumor-specific delivery to minimize toxic off-tumor effects. We conclude with a commentary on key challenges associated with these delivery methods and potential strategies to mitigate them.
Collapse
Affiliation(s)
- Ryunosuke Hoshi
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Kristyna A. Gorospe
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Hagar I. Labouta
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Leslie Dan Faculty of Pharmacy, University of Toronto, St. George Campus, Toronto, ON M5S 3M2, Canada
- Biomedical Engineering, Faculty of Applied Science and Engineering, University of Toronto, St. George Campus, Toronto, ON M5S 3E2, Canada
| | - Taha Azad
- Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Health Campus, Sherbrooke, QC J1K 2R1, Canada;
- Research Center, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC J1J 3H5, Canada
| | - Warren L. Lee
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Biochemistry, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada
- Medicine and the Interdepartmental Division of Critical Care Medicine, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5B 1T8, Canada
| | - Kelsie L. Thu
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| |
Collapse
|
13
|
Ohlinger J, Vordermark D, Ostheimer C, Bache M, Tzschoppe T, Demircan K, Schomburg L, Medenwald D, Seliger B. Change in the serum selenium level of patients with non-metastatic and metastatic non-small cell lung cancer (NSCLC) during radiotherapy as a predictive factor for survival. Strahlenther Onkol 2024:10.1007/s00066-024-02276-w. [PMID: 39240366 DOI: 10.1007/s00066-024-02276-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/07/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Lung cancer remains a serious medical problem. The trace element selenium seems to be a promising prognostic marker or therapeutic option for cancer patients. METHODS We enrolled 99 patients with histologically confirmed NSCLC undergoing radiotherapy. The serum selenium level of these patients was determined prior to irradiation (t0), after reaching 20 Gy (t1), and at the end of radiotherapy (t2). Selenium concentrations were measured with total-reflection X‑ray fluorescence (TXRF) spectroscopy. We formed three subgroups according to the change in serum selenium levels across timepoints, and Kaplan-Meier analysis was used to estimate overall survival (OS). Further subgroups were patients with/without metastatic disease. We used adjusted Cox regression models. RESULTS The change in selenium concentration was especially significant between t0 and t1 for the whole study group (hazard ratio [HR] = 0.5, p = 0.03) as well as in patients with metastasized NSCLC (HR = 0.3, p = 0.04) after adjustment. The baseline selenium value in patients with non-metastasized NSCLC was associated with overall survival (HR = 0.3, p = 0.04). The change in selenium levels between t0 and t2 was significant in patients with metastatic lung cancer (HR = 0.1, p = 0.03). Patients with increased serum selenium levels during radiotherapy between the start of treatment (t0) and t1 had better OS (HR = 0.46, p = 0.05). CONCLUSION Especially patients with increasing selenium levels during radiotherapy showed an improved overall survival. Thus, serum selenium might be a predictive factor for OS in NSCLC patients. The value of supplementation of the trace element is subject to future research.
Collapse
Affiliation(s)
- Julia Ohlinger
- Medical Faculty, Radiation Therapy Clinic, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Germany
| | - Dirk Vordermark
- Medical Faculty, Radiation Therapy Clinic, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Germany
| | - Christian Ostheimer
- Medical Faculty, Radiation Therapy Clinic, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Germany
| | - Matthias Bache
- Medical Faculty, Radiation Therapy Clinic, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Germany
| | - Therese Tzschoppe
- Medical Faculty, Radiation Therapy Clinic, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Germany
| | - Kamil Demircan
- Charité-University Medicine Berlin, Institute for Experimental Endocrinology, Berlin, Germany
| | - Lutz Schomburg
- Charité-University Medicine Berlin, Institute for Experimental Endocrinology, Berlin, Germany
| | - Daniel Medenwald
- Medical Faculty, Radiation Therapy Clinic, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Germany.
| | - Barbara Seliger
- Medical Faculty, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Institute for Translational Immunology, Brandenburg Medical School "Theodor Fontane", Brandenburg, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
14
|
Li H, Wang L, Ruan Z, Li X, Yang Y, Fang J, Wang R. CSE1L as a prognostic biomarker associated with pan cancer immune infiltration and drug sensitivity. Expert Rev Clin Immunol 2024; 20:1113-1125. [PMID: 38752776 DOI: 10.1080/1744666x.2024.2356747] [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: 11/06/2023] [Accepted: 05/13/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND Rising cancer-related mortality underscores the importance of biomarkers for treatment and prognosis, with Chromosome Segregation 1 Like (CSE1L) linked to various cancers yet its roles remain partially understood. This study investigates CSE1L's expression and oncogenic mechanisms in solid tumors. RESEARCH DESIGN AND METHODS We analyzed multi-omics data from 31 solid tumors, measured CSE1L in 41 head and neck carcinoma patients post-chemotherapy via qRT-PCR, and evaluated the impact of CSE1L knockdown on cell proliferation in A549 and HepG2 cells. RESULTS In this study, we observed significantly elevated levels of CSE1L RNA in 13 tumor tissues and protein levels in 8 tumor tissues compared to their corresponding adjacent normal tissues. Additionally, our investigation unveiled a correlation between heightened CSE1L expression in tumor tissues and worsened patient prognosis, poor response to immunotherapy, and diminished effectiveness of neoadjuvant chemotherapy. Through an analysis of CSE1L mechanisms, we discovered its potential involvement in promoting tumor cell proliferation, enhancing drug resistance, and influencing immune infiltration, thereby impacting patient prognosis and treatment outcomes. Finally, we delved into the potential mechanisms underlying upregulation of CSE1L in tumor tissues. CONCLUSION Our findings demonstrate that CSE1L promotes tumor development in various malignancies, highlighting its potential as both a therapeutic target and prognostic indicator.
Collapse
Affiliation(s)
- Haiyang Li
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lingwa Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhaohui Ruan
- Department of Major Emerging Infectious Diseases, Changping Laboratory, Beijing, China
| | - Xiaoyan Li
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha, China
| | - Yifan Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jugao Fang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ru Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Xu S, Xu Y, Solek NC, Chen J, Gong F, Varley AJ, Golubovic A, Pan A, Dong S, Zheng G, Li B. Tumor-Tailored Ionizable Lipid Nanoparticles Facilitate IL-12 Circular RNA Delivery for Enhanced Lung Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400307. [PMID: 38657273 DOI: 10.1002/adma.202400307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/24/2024] [Indexed: 04/26/2024]
Abstract
The advancement of message RNA (mRNA) -based immunotherapies for cancer is highly dependent on the effective delivery of RNA (Ribonucleic) payloads using ionizable lipid nanoparticles (LNPs). However, the clinical application of these therapies is hindered by variable mRNA expression among different cancer types and the risk of systemic toxicity. The transient expression profile of mRNA further complicates this issue, necessitating frequent dosing and thus increasing the potential for adverse effects. Addressing these challenges, a high-throughput combinatorial method is utilized to synthesize and screen LNPs that efficiently deliver circular RNA (circRNA) to lung tumors. The lead LNP, H1L1A1B3, demonstrates a fourfold increase in circRNA transfection efficiency in lung cancer cells over ALC-0315, the industry-standard LNPs, while providing potent immune activation. A single intratumoral injection of H1L1A1B3 LNPs, loaded with circRNA encoding interleukin-12 (IL-12), induces a robust immune response in a Lewis lung carcinoma model, leading to marked tumor regression. Immunological profiling of treated tumors reveals substantial increments in CD45+ leukocytes and enhances infiltration of CD8+ T cells, underscoring the ability of H1L1A1B3 LNPs to modulate the tumor microenvironment favorably. These results highlight the potential of tailored LNP platforms to advance RNA drug delivery for cancer therapy, broadening the prospects for RNA immunotherapeutics.
Collapse
Affiliation(s)
- Shufen Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Yue Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Nicholas C Solek
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Jingan Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Fanglin Gong
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Andrew James Varley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Alex Golubovic
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Anni Pan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Songtao Dong
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Gang Zheng
- Institute of Medical Science, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, M5G 2C1, Canada
| |
Collapse
|
16
|
Dunne EG, Fick CN, Isbell JM, Chaft JE, Altorki N, Park BJ, Spicer J, Forde PM, Gomez D, Iyengar P, Harpole DH, Stinchcombe TE, Liberman M, Bott MJ, Adusumilli PS, Huang J, Rocco G, Jones DR. The Emerging Role of Immunotherapy in Resectable Non-Small Cell Lung Cancer. Ann Thorac Surg 2024; 118:119-129. [PMID: 38316378 PMCID: PMC11194155 DOI: 10.1016/j.athoracsur.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Despite surgical resection, long-term survival of patients with resectable non-small cell lung cancer (NSCLC) remains poor. Adjuvant chemotherapy, the standard of care for locally advanced NSCLC, provides a marginal 5.4% benefit in survival. Immune checkpoint inhibitors (ICIs) have shown a significant survival benefit in some patients with advanced NSCLC and are being evaluated for perioperative use in resectable NSCLC. METHODS We conducted a literature search using the PubMed online database to identify clinical trials of immunotherapy in resectable NSCLC and studies analyzing biomarkers and immune priming strategies. RESULTS Building on previous phase I and II trials, randomized phase III trials have shown efficacy of neoadjuvant nivolumab, perioperative pembrolizumab, adjuvant atezolizumab, and adjuvant pembrolizumab in the treatment of NSCLC with improvement of event-free/disease-free survival of 24% to 42%, leading to United States Food and Drug Administration approval of these drugs in the treatment of resectable NSCLC. Three additional phase III trials have also recently reported the use of immunotherapy both before and after surgery, with pathologic complete response rates of 17% to 25%, significantly better than chemotherapy alone. Perioperative ICI therapy has comparable perioperative morbidity to chemotherapy alone and does not impair surgical outcomes. CONCLUSIONS Perioperative immunotherapy, in combination with chemotherapy, is safe and improves outcomes in patients with resectable NSCLC. Questions regarding patient selection, the need for adjuvant ICI therapy after neoadjuvant chemoimmunotherapy, and the duration of perioperative immunotherapy remain to be answered by future trials.
Collapse
Affiliation(s)
- Elizabeth G Dunne
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cameron N Fick
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James M Isbell
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jamie E Chaft
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nasser Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, New York
| | - Bernard J Park
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan Spicer
- Department of Thoracic Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Patrick M Forde
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Daniel Gomez
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Puneeth Iyengar
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H Harpole
- Division of Cardiovascular and Thoracic Surgery, Duke University Medical Center, Durham, North Carolina
| | - Thomas E Stinchcombe
- Division of Medical Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Moishe Liberman
- Division of Thoracic Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Matthew J Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James Huang
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gaetano Rocco
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
17
|
Sakaeda K, Kurose K, Matsumura Y, Muto S, Fukuda M, Sugasaki N, Fukuda M, Takemoto S, Taniguchi H, Masuda T, Shimizu K, Kataoka Y, Irino Y, Sakai Y, Atarashi Y, Yanagida M, Hattori N, Mukae H, Nakata M, Kanda E, Oga T, Suzuki H, Oka M. Automated immunoassay of serum NY-ESO-1 and XAGE1 antibodies for predicting clinical benefit with immune checkpoint inhibitor (ICI) in advanced non-small cell lung cancer. Cancer Treat Res Commun 2024; 40:100830. [PMID: 38964205 DOI: 10.1016/j.ctarc.2024.100830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND NY-ESO-1 and XAGE1 cancer/testis antigens elicit humoral and cellular immune responses in NSCLC patients. We aimed to predict clinical benefit with ICI monotherapy, using an automated immunoassay of NY-ESO-1/XAGE1 antibodies (Abs). METHODS This study enrolled 99 NSCLC patients who received nivolumab after chemotherapy, including 21 patients harboring EGFR, ALK, or KRAS alterations. The cutoff value (10 units/mL) of NY-ESO-1 and XAGE1 Ab was determined based on Ab levels in non-malignant controls, and NY-ESO-1/XAGE1 Abs in NSCLC were measured before nivolumab. Differences in PFS and OS between the Ab-positive and Ab-negative groups were retrospectively analyzed using Cox regression analysis after applying inverse probability of treatment weighting (IPTW). RESULTS NY-ESO-1/XAGE1 Abs were positive in 28 NSCLC, who responded more highly to nivolumab than the Ab-negatives (response rate 50.0% vs. 15.5 %, p < 0.0007). The IPTW-adjusted positives and negatives for NY-ESO-1/XAGE1 Abs were 24.5 and 70.2, respectively. The Ab-positives showed longer IPTW-adjusted PFS (HR = 0.59, 95 % CI: 0.39-0.90, p = 0.014) and IPTW-adjusted OS (HR = 0.51, 95 % CI: 0.32-0.81, p = 0.004) than the Ab-negatives. Among NSCLC harboring driver genes, the Ab-positives (n = 10) showed longer PFS (HR = 0.34, 95 % CI: 0.13-0.89, p = 0.029) and OS (HR = 0.27, 95 % CI: 0.098-0.75, p = 0.012) than the Ab-negatives (n = 11). CONCLUSION Our immunoassay of NY-ESO-1/XAGE1 Abs is probably useful for predicting the clinical benefit with nivolumab in NSCLC, including those harboring driver genes. These results suggest that our immunoassay may be useful in ICI monotherapy for NSCLC.
Collapse
Affiliation(s)
- Kanako Sakaeda
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo 651-0073, Japan
| | - Koji Kurose
- Respiratory Medicine, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Yuki Matsumura
- Thoracic Surgery, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Satoshi Muto
- Thoracic Surgery, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Minoru Fukuda
- Respiratory Medicine, Nagasaki Prefecture Shimabara Hospital, Shimabara, Nagasaki 855-0861, Japan
| | - Nanae Sugasaki
- Respiratory Medicine, Nagasaki Prefecture Shimabara Hospital, Shimabara, Nagasaki 855-0861, Japan
| | - Masaaki Fukuda
- Respiratory Medicine, The Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Nagasaki 852-8511, Japan
| | - Shinnosuke Takemoto
- Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Nagasaki 852-8501, Japan
| | - Hirokazu Taniguchi
- Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Nagasaki 852-8501, Japan
| | - Takeshi Masuda
- Respiratory Internal Medicine, Hiroshima University Hospital, Hiroshima, Hiroshima 734-8551, Japan
| | - Katsuhiko Shimizu
- Thoracic Surgery, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Yuki Kataoka
- Scientific Research Works Peer Support Group (SRWS-PSG), Osaka, Japan
| | - Yasuhiro Irino
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo 651-0073, Japan
| | - Yumiko Sakai
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo 651-0073, Japan
| | - Yusuke Atarashi
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo 651-0073, Japan
| | - Masatoshi Yanagida
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo 651-0073, Japan
| | - Noboru Hattori
- Respiratory Internal Medicine, Hiroshima University Hospital, Hiroshima, Hiroshima 734-8551, Japan
| | - Hiroshi Mukae
- Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Nagasaki 852-8501, Japan
| | - Masao Nakata
- Thoracic Surgery, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Eiichiro Kanda
- Medical Science, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Toru Oga
- Respiratory Medicine, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Hiroyuki Suzuki
- Thoracic Surgery, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Mikio Oka
- Immuno-Oncology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan.
| |
Collapse
|
18
|
Matsui T, Taniguchi S, Ishii M. Function of alveolar macrophages in lung cancer microenvironment. Inflamm Regen 2024; 44:23. [PMID: 38720352 PMCID: PMC11077793 DOI: 10.1186/s41232-024-00335-4] [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: 03/16/2024] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Cancer tissues contain a wide variety of immune cells that play critical roles in suppressing or promoting tumor progression. Macrophages are one of the most predominant populations in the tumor microenvironment and are composed of two classes: infiltrating macrophages from the bone marrow and tissue-resident macrophages (TRMs). This review aimed to outline the function of TRMs in the tumor microenvironment, focusing on lung cancer. REVIEW Although the functions of infiltrating macrophages and tumor-associated macrophages have been intensively analyzed, a comprehensive understanding of TRM function in cancer is relatively insufficient because it differs depending on the tissue and organ. Alveolar macrophages (AMs), one of the most important TRMs in the lungs, are replenished in situ, independent of hematopoietic stem cells in the bone marrow, and are abundant in lung cancer tissue. Recently, we reported that AMs support cancer cell proliferation and contribute to unfavorable outcomes. CONCLUSION In this review, we introduce the functions of AMs in lung cancer and their underlying molecular mechanisms. A thorough understanding of the functions of AMs in lung cancer will lead to improved treatment outcomes.
Collapse
Affiliation(s)
- Takahiro Matsui
- Department of Immunology and Cell Biology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Seiji Taniguchi
- Department of Immunology and Cell Biology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Thoracic Surgery, Osaka Habikino Medical Center, Habikino, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
19
|
Zhang B, Chen J, Yu H, Li M, Cai M, Chen L. Prognostic Nutritional Index Predicts Efficacy and Immune-Related Adverse Events of First-Line Chemoimmunotherapy in Patients with Extensive-Stage Small-Cell Lung Cancer. J Inflamm Res 2024; 17:1777-1788. [PMID: 38523686 PMCID: PMC10959246 DOI: 10.2147/jir.s450804] [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: 11/21/2023] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
Background Currently, there is a lack of well-established markers to predict the efficacy of chemoimmunotherapy in small-cell lung cancer (SCLC). Neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), advanced lung cancer inflammation index (ALI) and prognostic nutritional index (PNI) are associated with prognosis in several tumors, whereas their predictive role in SCLC remains unclear. Methods A retrospective study was conducted at Sun Yat-sen University Cancer Center, involving extensive-stage SCLC (ES-SCLC) patients who received first-line chemoimmunotherapy between January 2020 and December 2021. Peripheral blood biomarkers were extracted from medical records and their correlation with prognosis and immune-related adverse events (IRAEs) was analyzed. Results A total of 114 patients were included. Patients with a low PLR, high ALI and high PNI had prolonged progression-free survival (PFS) compared to those with a high PLR, low ALI and low PNI. Patients with a low NLR, low PLR, high ALI and high PNI had prolonged overall survival (OS) compared to those with a high NLR, high PLR, low ALI and low PNI. Cox regression model showed that PNI was an independent risk factor for both PFS and OS. ROC curve showed that PNI outperforms NLR, PLR and ALI in predicting both PFS and OS. The PNI-based nomogram demonstrated strong predictive capability for both PFS and OS. In addition, there was a significant correlation between PNI and IRAEs. Conclusion A high baseline PNI might be associated with improved prognosis and the occurrence of IRAEs in ES-SCLC patients treated with first-line chemoimmunotherapy.
Collapse
Affiliation(s)
- Baishen Zhang
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Jing Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Hui Yu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Meichen Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Muyan Cai
- Department of Pathology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Likun Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| |
Collapse
|
20
|
Li J, Li Z, Yang W, Pan J, You H, Yang L, Zhang X. Development and verification of a novel immunogenic cell death-related signature for predicting the prognosis and immune infiltration in triple-negative breast cancer. Cancer Rep (Hoboken) 2024; 7:e2007. [PMID: 38425247 PMCID: PMC10905160 DOI: 10.1002/cnr2.2007] [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: 09/26/2023] [Revised: 01/01/2024] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Insufficient understanding of the pathogenesis and tumor immunology of triple-negative breast cancer (TNBC) has limited the development of immunotherapy. The importance of tumor microenvironment (TME) in immunotyping, prognostic assessment and immunotherapy efficacy of cancer has been emphasized, however, potential immunogenic cell death (ICD) related genes function in TME of TNBC has been rarely investigated. AIMS To initially explore the role and related mechanisms of ICD in TNBC, especially the role played in the TME of TNBC, and to identify different relevant subtypes based on ICD, and then develop an ICD-related risk score to predict each TNBC patient TME status, prognosis and immunotherapy response. METHODS AND RESULTS In this study, we identified distinct ICD-related modification patterns based on 158 TNBC cases in the TCGA-TNBC cohort. We then investigated the possible correlation between ICD-related modification patterns and TME cell infiltration characteristics in TNBC. By using univariate Cox and least absolute shrinkage and selection operator (LASSO) regression analysis, we created a risk scoring system (ICD score) to quantifiably evaluate the impact of ICD-related modification patterns in individual TNBC patient. Two different ICD-related modification patterns were found with significant differences in immune infiltration. Lower ICD score was correlated with higher immune infiltration, tumor mutational burden and significantly enriched in immune-related pathways, indicating a strong ability to activate immune response, which might account for relatively favorable prognosis of TNBC patients and could serve as a predictor to select suitable candidates for immunotherapy. We used two independent cohorts, GSE58812 cohort and Metabric cohort to validate prognosis and immunohistochemistry for preliminary in vitro validation. CONCLUSION This study evidenced that the ICD-related modification patterns might exert pivotal roles in the immune infiltration landscape of TNBC and ICD score might act as potential predictors of prognostic assessment and immunotherapy response. This research provides unique insights for individualize immune treatment strategies and promising immunotherapy candidates screening.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Gastrointestinal and Gland SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Zhengtian Li
- Department of Bone and Joint SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Wenkang Yang
- Department of Gastrointestinal and Gland SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jianmin Pan
- Department of Gastrointestinal and Gland SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Huazong You
- Department of Gastrointestinal and Gland SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Lixiang Yang
- Department of Gastrointestinal and Gland SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Xiaodong Zhang
- Department of Gastrointestinal and Gland SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
21
|
Wang Z, Yao X, Wang K, Wang B. TFR1-Mediated Iron Metabolism Orchestrates Tumor Ferroptosis and Immunity in Non-Small Cell Lung Cancer. J Environ Pathol Toxicol Oncol 2024; 43:1-12. [PMID: 38505909 DOI: 10.1615/jenvironpatholtoxicoloncol.2023049084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
This study aimed to investigate the underlying molecular mechanisms of transferrin receptor (TFR1) in non-small cell lung cancer (NSCLC). Histological analysis was performed using hematoxylin-eosin (HE) staining. The number of CD8+ T cell were determined by flow cytometry and immunofluorescence assays. mRNA levels were analyzed by qRT-PCR. Protein expression was detected by western blot. Ferroptosis was detected by using propidium iodide (PI) staining. Xenograft experiment was applied for determining tumor growth. The results showed that interferon (IFN)-γ plus iron dextran (FeDx) induced iron overload and the ferroptosis of NSCLC cells. Moreover, IFN-γ-mediated upregulation of TFR1 promoted ferritinophagy and tumor cell ferroptosis via blocking via blocking ferritin heavy chain 1 (FTH1)/ ferritin light chain (FTL) signaling. However, TFR1 knockout suppressed the ferroptosis of tumor cells. Furthermore, FeDx-mediated iron overload promoted the sensitivity of anti-programmed death ligand 1 (PD-L1) therapies. Clinically, TFR1 was downregulated in NSCLC patients. Low levels of TFR1 predicted decreased CD8+ T cells. Taken together, IFN-γ combined with iron metabolism therapies may provide a novel alternative for NSCLC.
Collapse
Affiliation(s)
- Zunqiao Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing 210029, P.R China
| | - Xingkai Yao
- Department of Thoracic Surgery, People's Hospital of Luhe District in Nanjing, Nanjing 210000, P.R China
| | - Keping Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing 210029, P.R China
| | - Bin Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing 210029, P.R China
| |
Collapse
|
22
|
Lasala R, Zovi A, Isgrò V, Romagnoli A, Musicco F, Santoleri F. Time to treatment discontinuation in first-line non-small cell lung carcinoma: an overview. Curr Med Res Opin 2023; 39:1603-1612. [PMID: 36932463 DOI: 10.1080/03007995.2023.2192610] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVE Time To Discontinuation (TTD) is defined as the time from the start of treatment to the end of treatment, usually occurring due to loss of efficacy or occurrence of adverse events. It has become an important surrogate efficacy endpoint especially in real-world studies due to its correlation with endpoints such as Progression Free Survival (PFS). The aim of the study is to conduct a literature review of all studies reporting TTD in first-line therapy of Non-Small Cell Lung Cancer (NSCLC). METHODS All articles that reported TTD for any first-line treatment of NSCLC as of 30 June 2022 were extracted from the PubMed search engine. From these articles, the drugs, study type, and TTD values were extracted. A descriptive analysis of the studies was made, dividing the TTD by subgroup according to the type of treatment (traditional chemotherapy, target therapy, immunotherapy) and study design (clinical trials, real world studies). RESULTS Fifty-five studies were considered for the analysis, of which 12 were published in 2021; 28 were clinical trials and 27 were real-world studies. Thirty of the studies considered involved conventional chemotherapy and expressed TTD values from 1.4 to 4.5 months, 5 of the studies considered involved immunotherapy with TTD values from 2.1 to 7.4 months and 18 of the studies considered target therapy, with TTD values from 4 to 31 months. The clinical trials reported TTD values from 1.4 to 16 months and the real-world studies from 2 to 31 months. CONCLUSION Studies reporting TTD are increasing, most notably real-world studies. Given the increasing importance of TTD as an efficacy endpoint, it becomes critical to measure and monitor it in various therapeutic settings such as NSCLC. This is the first study to review all TTD values of drugs used in first-line NSCLC.
Collapse
Affiliation(s)
- Ruggero Lasala
- Hospital Pharmacy of Corato, Local Health Unit of Bari, Italy
| | - Andrea Zovi
- Ministry of Health, Ministero della Salute, Rome, Italy
| | - Valentina Isgrò
- Clinical Pharmacy, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Alessia Romagnoli
- Territorial Pharmaceutical Service, Local Health Unit of Lanciano Vasto Chieti, Chieti, Italy
| | - Felice Musicco
- Hospital Pharmacy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | |
Collapse
|
23
|
Janzic U, Shalata W, Szymczak K, Dziadziuszko R, Jakopovic M, Mountzios G, Płużański A, Araujo A, Charpidou A, Agbarya A. Real-World Experience in Treatment of Patients with Non-Small-Cell Lung Cancer with BRAF or cMET Exon 14 Skipping Mutations. Int J Mol Sci 2023; 24:12840. [PMID: 37629023 PMCID: PMC10454089 DOI: 10.3390/ijms241612840] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
BRAF and cMET exon 14 skipping are rare mutations of NSCLC. The treatment sequence in these cases for the first and second line is not clear. An international registry was created for patients with advanced NSCLC harboring BRAF or cMET exon 14 skipping mutations, diagnosed from January 2017 to June 2022. Clinicopathological and molecular data and treatment patterns were recorded. Data on 58 patients, from eight centers across five countries, were included in the final analysis. We found that 40 patients had the cMET exon 14 skipping mutation and 18 had the BRAF V600E mutation. In total, 53 and 28 patients received first- and second-line treatments, respectively, among which 52.8% received targeted therapy (TT) in the first line and 53.5% in the second line. The overall response rate (ORR) and disease control rate (DCR) for first-line treatment with TT vs. other treatment such as immune checkpoint inhibitors ± chemotherapy (IO ± CT) were 55.6% vs. 21.7% (p = 0.0084) and 66.7% vs. 39.1% (p = 0.04), respectively. The type of treatment in first-line TT vs. other affected time to treatment discontinuation (TTD) was 11.6 m vs. 4.6 m (p= 0.006). The overall survival for the whole group was 15.4 m and was not statistically affected by the type of treatment (19.2 m vs. 13.5 m; p = 0.83).
Collapse
Affiliation(s)
- Urska Janzic
- Department of Medical Oncology, University Clinic Golnik, 4204 Golnik, Slovenia
- Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Walid Shalata
- The Legacy Heritage Cancer Center & Dr. Larry Norton Institute, Soroka Medical Center, Ben Gurion University, Beer Sheva 84105, Israel
| | - Katarzyna Szymczak
- Department of Oncology and Radiotherapy and Early Phase Clinical Trials Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Rafał Dziadziuszko
- Department of Oncology and Radiotherapy and Early Phase Clinical Trials Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Marko Jakopovic
- Department for Respiratory Diseases Jordanovac, University Hospital Centre Zagreb, 10 000 Zagreb, Croatia
| | - Giannis Mountzios
- Clinical Trials Unit, Fourth Oncology Department, Henry Dunant Hospital Center, 115 26 Athens, Greece
| | - Adam Płużański
- Department of Lung Cancer and Chest Tumours, The Maria Skłodowska-Curie National Research Institute of Oncology, 00-001 Warsaw, Poland
| | - Antonio Araujo
- Department of Medical Oncology, CHUPorto—University Hospitalar Center of Porto, 4099-001 Porto, Portugal
| | - Andriani Charpidou
- Oncology Unit, 3rd Department of Medicine, “Sotiria” Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 106 79 Athens, Greece
| | - Abed Agbarya
- Department of Oncology, Bnai-Zion Medical Center, 47 Golomb Avenue, Haifa 31048, Israel
| |
Collapse
|
24
|
Xiao H, Liu Q, Li L. MFMANet: Multi-feature Multi-attention Network for efficient subtype classification on non-small cell lung cancer CT images. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2023.104768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
25
|
Pan Y, Gao J, Lin J, Ma Y, Hou Z, Lin Y, Wen S, Pan M, Lu F, Huang H. High-dimensional single-cell analysis unveils distinct immune signatures of peripheral blood in patients with pancreatic ductal adenocarcinoma. Front Endocrinol (Lausanne) 2023; 14:1181538. [PMID: 37347110 PMCID: PMC10281055 DOI: 10.3389/fendo.2023.1181538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with poor response to immune checkpoint inhibitors. The mechanism of such poor response is not completely understood. Methods We assessed T-cell receptor (TCR) repertoire and RNA expression at the single-cell level using high-dimensional sequencing of peripheral blood immune cells isolated from PDAC patients and from healthy human controls. We validated RNA-sequencing data by performing mass cytometry (CyTOF) and by measuring serum levels of multiple immune checkpoint proteins. Results We found that proportions of T cells (CD45+CD3+) were decreased in PDAC patients compared to healthy controls, while proportion of myeloid cells was increased. The proportion of cytotoxic CD8+ T cells and the level of cytotoxicity per cell were increased in PDAC patients, with reduced TCR clonal diversity. We also found a significantly enriched S100A9+ monocyte population and an increased level of TIM-3 expression in immune cells of peripheral blood in PDAC patients. In addition, the serum level of soluble TIM-3 (sTIM-3) was significantly higher in PDAC patients compared to the non-PDAC participants and correlated with worse survival in two independent PDAC cohorts. Moreover, sTIM-3 exhibited a valuable role in diagnosis of PDAC, with sensitivity and specificity of about 80% in the training and validation groups, respectively. We further established an integrated model by combining sTIM-3 and carbohydrate antigen 19- 9 (CA19-9), which had an area under the curve of 0.974 and 0.992 in training and validation cohorts, respectively. Conclusion Our RNA-seq and proteomic results provide valuable insight for understanding the immune cell composition of peripheral blood of patients with PDAC.
Collapse
Affiliation(s)
- Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianfeng Gao
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiajing Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuan Ma
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zelin Hou
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yali Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shi Wen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Minggui Pan
- Department of Oncology and Hematology and Division of Research, Kaiser Permanente, Santa Clara, CA, United States
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
26
|
Wei S, Xing J, Lu K, Wang K, Yu W. NPM3 as a novel oncogenic factor and poor prognostic marker contributes to cell proliferation and migration in lung adenocarcinoma. Hereditas 2023; 160:27. [PMID: 37254219 PMCID: PMC10230701 DOI: 10.1186/s41065-023-00289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/21/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide, and despite recent advances in targeted therapies and immunotherapies, the clinical benefit remains limited. Therefore, there is an urgent need to further investigate the molecular mechanisms underlying lung cancer. The aim of this study was to investigate the expression and function of NPM3 in the tumor microenvironment of lung adenocarcinoma (LUAD). METHODS We utilized bioinformatics tools and databases, including UALCAN, GEPIA2, HPA, and Sangerbox, to analyze NPM3 expression in LUAD samples and its association with prognosis and mutational landscape. NPM3 expression in various cell types was assessed at the single cell level using the TISCH database. We also used algorithms such as TIMER and EPIC to explore the crosstalk between NPM3 expression and immune features. KEGG enrichment analysis was performed to identify potential signaling pathways of NPM3. Finally, we employed siRNA knockdown strategy to investigate the effect of NPM3 on LUAD cell proliferation and migration in vitro. RESULTS NPM3 was significantly upregulated in LUAD tissues and was strongly associated with poor prognosis and TP53 gene mutations. Single-cell sequencing analysis revealed that NPM3 was expressed in immune cells (dendritic cells and monocytes/macrophages) in the tumor microenvironment. Moreover, NPM3 expression was negatively associated with immune B cell and CD4 T cell infiltration, as well as with several immune-related genes (including CCL22, CXCR2, CX3CR1, CCR6, HLA-DOA, HLA-DQA2). KEGG enrichment analysis indicated that NPM3 expression was associated with cell cycle, CAMs, and NSCLC pathway genes. Finally, in vitro experiments showed that NPM3 knockdown inhibited LUAD cell proliferation and migration in NCI-H1299 and SPC-A1 cells, and suppressed the expression of CCNA2 and MAD2L1. CONCLUSION Elevated NPM3 expression predicts poor clinical outcome and an immunosuppressive microenvironment in LUAD tissues. NPM3 promotes LUAD progression by promoting cell proliferation and migration, and targeting NPM3 may represent a novel therapeutic strategy for LUAD.
Collapse
Affiliation(s)
- Shan Wei
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, Zhejiang, People's Republic of China
- Department of Respiratory and Critical Care Medicine, The Affiliated People's Hospital of Ningbo University (Ningbo Yinzhou People's Hospital), No.251, Baizhang Road, Ningbo, 315040, Zhejiang, People's Republic of China
| | - Jing Xing
- Department of Respiratory and Critical Care Medicine, The Affiliated People's Hospital of Ningbo University (Ningbo Yinzhou People's Hospital), No.251, Baizhang Road, Ningbo, 315040, Zhejiang, People's Republic of China
- Ningbo University School of Medicine, Zhejiang Province, Ningbo, People's Republic of China
| | - Kaining Lu
- Department of Urology, The Affiliated First Hospital of Ningbo University (Ningbo First Hospital), No.59, Liuting Street, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, Zhejiang, People's Republic of China
| | - Wanjun Yu
- Department of Respiratory and Critical Care Medicine, The Affiliated People's Hospital of Ningbo University (Ningbo Yinzhou People's Hospital), No.251, Baizhang Road, Ningbo, 315040, Zhejiang, People's Republic of China.
| |
Collapse
|
27
|
Ahvonen J, Luukkaala T, Laitinen T, Jukkola A. Survival with lung cancer in Finland has not improved during 2007-2019-a single center retrospective population-based real-world study. Acta Oncol 2023:1-8. [PMID: 37257498 DOI: 10.1080/0284186x.2023.2213444] [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: 05/30/2022] [Accepted: 05/09/2023] [Indexed: 06/02/2023]
Abstract
OBJECTIVES According to the CONCORD-3 study, the 5-year survival rate of lung cancer patients in Finland has not improved during the twenty-first century. In the present study, we evaluated the survival trends of lung cancer patients diagnosed and treated in one of the five university hospitals in Finland to determine possible explanatory factors behind the lack of improved survival. MATERIAL AND METHODS This retrospective population-based study included all lung cancer patients diagnosed in Tampere University Hospital in 2007-2019 (N = 3041). The study population was divided into two subcohorts: the patients diagnosed in 2007-2012 and those diagnosed in 2013-2019. The two subcohorts were then compared to analyze the temporal changes in survival and the distribution of prognostic factors. RESULTS A comparison of the patients diagnosed in 2007-2012 and 2013-2019 showed that the patients' overall survival had remained unchanged. The median overall survival was 8.7 months in the earlier subcohort and 9.2 months in the later subcohort. The respective 5-year survival rates were 16.6% and 17.8%, and these differences were not statistically significant. The proportion of stage IV patients (approximately 59% in both subcohorts) and their risk of death were similar for the two subcohorts. According to the regression analysis, male gender, advanced stage, and poor Eastern Cooperative Oncology Group performance status were independent risk factors for death, while a never-smoking status and mutation-positive disease were associated with a decreased risk of death, but only in the later cohort. CONCLUSION Echoing the results of CONCORD-3, this study confirmed that the real-world survival of unselected lung cancer populations in Finland has not improved over the last 15 years, mainly because of the unchanged proportions of patients with late-stage lung cancer. This calls for earlier recognition of lung cancer, achieved by screening and increasing awareness of the disease.
Collapse
Affiliation(s)
- Jarkko Ahvonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Tiina Luukkaala
- Research, Development and Innovation Center, Tampere University Hospital, Tampere, Finland
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Tarja Laitinen
- Administration Center, Tampere University Hospital, Tampere, Finland
| | - Arja Jukkola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
28
|
Shen L, Brown JR, Johnston SA, Altan M, Sykes KF. Predicting response and toxicity to immune checkpoint inhibitors in lung cancer using antibodies to frameshift neoantigens. J Transl Med 2023; 21:338. [PMID: 37217961 DOI: 10.1186/s12967-023-04172-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
PURPOSE To evaluate a new class of blood-based biomarkers, anti-frameshift peptide antibodies, for predicting both tumor responses and adverse immune events to immune checkpoint inhibitor (ICI) therapies in advanced lung cancer patients. EXPERIMENTAL DESIGN Serum samples were obtained from 74 lung cancer patients prior to palliative PD-(L)1 therapies with subsequently recorded tumor responses and immune adverse events (irAEs). Pretreatment samples were assayed on microarrays of frameshift peptides (FSPs), representing ~ 375,000 variant peptides that tumor cells can be informatically predicted to produce from translated mRNA processing errors. Serum-antibodies specifically recognizing these ligands were measured. Binding activities preferentially associated with best-response and adverse-event outcomes were determined. These antibody bound FSPs were used in iterative resampling analyses to develop predictive models of tumor response and immune toxicity. RESULTS Lung cancer serum samples were classified based on predictive models of ICI treatment outcomes. Disease progression was predicted pretreatment with ~ 98% accuracy in the full cohort of all response categories, though ~ 30% of the samples were indeterminate. This model was built with a heterogeneous sample cohort from patients that (i) would show either clear response or stable outcomes, (ii) would be administered either single or combination therapies and (iii) were diagnosed with different lung cancer subtypes. Removing the stable disease, combination therapy or SCLC groups from model building increased the proportion of samples classified while performance remained high. Informatic analyses showed that several of the FSPs in the all-response model mapped to translations of variant mRNAs from the same genes. In the predictive model for treatment toxicities, binding to irAE-associated FSPs provided 90% accuracy pretreatment, with no indeterminates. Several of the classifying FSPs displayed sequence similarity to self-proteins. CONCLUSIONS Anti-FSP antibodies may serve as biomarkers for predicting ICI outcomes when tested against ligands corresponding to mRNA-error derived FSPs. Model performances suggest this approach might provide a single test to predict treatment response to ICI and identify patients at high risk for immunotherapy toxicities.
Collapse
Affiliation(s)
- Luhui Shen
- Calviri, Inc, 850 N 5th St., Phoenix, AZ, 85004, USA
| | | | | | - Mehmet Altan
- MD Anderson Cancer Center, Department of Thoracic-Head & Neck Medical Oncology, Division of Cancer Medicine, Houston, TX, USA
| | | |
Collapse
|
29
|
Wang Y, Huang S, Feng X, Xu W, Luo R, Zhu Z, Zeng Q, He Z. Advances in efficacy prediction and monitoring of neoadjuvant immunotherapy for non-small cell lung cancer. Front Oncol 2023; 13:1145128. [PMID: 37265800 PMCID: PMC10229830 DOI: 10.3389/fonc.2023.1145128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has become mainstream in the treatment of non-small cell lung cancer (NSCLC). The idea of harnessing the immune system to fight cancer is fast developing. Neoadjuvant treatment in NSCLC is undergoing unprecedented change. Chemo-immunotherapy combinations not only seem to achieve population-wide treating coverage irrespective of PD-L1 expression but also enable achieving a pathological complete response (pCR). Despite these recent advancements in neoadjuvant chemo-immunotherapy, not all patients respond favorably to treatment with ICIs plus chemo and may even suffer from severe immune-related adverse effects (irAEs). Similar to selection for target therapy, identifying patients most likely to benefit from chemo-immunotherapy may be valuable. Recently, several prognostic and predictive factors associated with the efficacy of neoadjuvant immunotherapy in NSCLC, such as tumor-intrinsic biomarkers, tumor microenvironment biomarkers, liquid biopsies, microbiota, metabolic profiles, and clinical characteristics, have been described. However, a specific and sensitive biomarker remains to be identified. Recently, the construction of prediction models for ICI therapy using novel tools, such as multi-omics factors, proteomic tests, host immune classifiers, and machine learning algorithms, has gained attention. In this review, we provide a comprehensive overview of the different positive prognostic and predictive factors in treating preoperative patients with ICIs, highlight the recent advances made in the efficacy prediction of neoadjuvant immunotherapy, and provide an outlook for joint predictors.
Collapse
Affiliation(s)
- Yunzhen Wang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sha Huang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangwei Feng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangjue Xu
- Department of Thoracic Surgery, Longyou County People’s Hospital, Longyou, China
| | - Raojun Luo
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyi Zhu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingxin Zeng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengfu He
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
30
|
Ilié M, Hofman V, Bontoux C, Goffinet S, Benzaquen J, Heeke S, Boutros J, Lassalle S, Long-Mira E, Zahaf K, Lalvée S, Lespinet-Fabre V, Bordone O, Tanga V, Gómez-Caro A, Cohen C, Berthet JP, Marquette CH, Hofman P. Lack of correlation between MET and PD-L1 expression in non-small cell lung cancer revealed by comparative study of matched biopsies and surgical resection samples. Lung Cancer 2023; 181:107230. [PMID: 37150140 DOI: 10.1016/j.lungcan.2023.107230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023]
Abstract
INTRODUCTION Both MET expression and the PD-L1 tumor proportion score (TPS) are companion diagnostics for treatment of advanced non-small cell lung carcinoma (aNSCLC) patients. We evaluated the rate of correlation between MET expression and the PD-L1 TPS in matched biopsies and surgically resected specimens from NSCLC patients. PATIENTS AND METHODS This retrospective analysis assessed the prevalence and correlation between MET expression (SP44 clone) and the PD-L1 TPS (22C3 clone) by immunohistochemistry together with molecular alterations determined by targeted next-generation sequencing in matched lung biopsy and surgically lung resected specimens from 70 patients with NSCLC. RESULTS The study found a significant correlation between the MET H-score in surgical samples and matched biopsies (P-value < 0.0001), as well as between the PD-L1 TPS in paired biopsies and surgical samples (P-value < 0.0001). However, there was no significant correlation between the MET H-score or expression subgroups and the PD-L1 TPS in both types of paired samples (P-value = 0.47, and P-value = 0.90). The MET H-score was significantly higher in adenocarcinoma compared to squamous cell carcinoma (P-value < 0.0001). A mutational analysis showed that the MET H-score was significantly higher in NSCLC cases with targetable molecular alterations (P-value = 0.0095), while no significant correlation was found for the PD-L1 TPS. CONCLUSIONS Our study found no significant correlation between PD-L1 and MET expression in samples from NSCLC patients, highlighting the importance of personalized treatment strategies based on individual expression profiles. These findings provide valuable insight into the development of effective immunotherapy and targeted therapy for NSCLC patients.
Collapse
Affiliation(s)
- Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Samantha Goffinet
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Jonathan Benzaquen
- FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France; Department of Pulmonary Medicine and Thoracic Oncology, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jacques Boutros
- FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Department of Pulmonary Medicine and Thoracic Oncology, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Virginie Tanga
- Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France
| | - Abel Gómez-Caro
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Charlotte Cohen
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Jean-Philippe Berthet
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Charles-Hugo Marquette
- FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France; Department of Pulmonary Medicine and Thoracic Oncology, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France.
| |
Collapse
|
31
|
Wei Y, Xu Y, Wang M. Immune checkpoint inhibitors for the treatment of non-small cell lung cancer brain metastases. Chin Med J (Engl) 2023:00029330-990000000-00586. [PMID: 37106555 DOI: 10.1097/cm9.0000000000002163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Indexed: 04/29/2023] Open
Abstract
ABSTRACT Lung cancer has the highest risk of brain metastasis (BM) among all solid carcinomas. The emergence of BM has a significant impact on the selection of oncologic treatment for patients. Immune checkpoint inhibitors (ICIs) are the most promising treatment option for patients without druggable mutations and have been shown to improve survival in patients with non-small cell lung cancer (NSCLC) BM in clinical trials with good safety. Moreover, ICI has shown certain effects in NSCLC BM, and the overall intracranial efficacy is comparable to extracranial efficacy. However, a proportion of patients showed discordant responses in primary and metastatic lesions, suggesting that multiple mechanisms may exist underlying ICI activity in BM. According to studies pertaining to tumor immune microenvironments, ICIs may be capable of provoking immunity in situ. Meanwhile, systematic immune cells activated by ICIs can migrate into the central nervous system and exert antitumor effects. This review summarizes the present evidence for ICI treatment efficacy in NSCLC BM and proposes the possible mechanisms of ICI treatment for NSCLC BMs based on existing evidence.
Collapse
Affiliation(s)
- Yuxi Wei
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Peking Union Medical College (PUMC) and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
32
|
Zhang J, Hao L, Qi M, Xu Q, Zhang N, Feng H, Shi G. Radiomics nomogram for preoperative differentiation of pulmonary mucinous adenocarcinoma from tuberculoma in solitary pulmonary solid nodules. BMC Cancer 2023; 23:261. [PMID: 36944978 PMCID: PMC10029225 DOI: 10.1186/s12885-023-10734-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023] Open
Abstract
OBJECTIVE To develop and validate predictive models using clinical parameters, radiomic features and a combination of both for preoperative differentiation of pulmonary nodular mucinous adenocarcinoma (PNMA) from pulmonary tuberculoma (PTB). METHOD A total of 124 and 53 patients with PNMA and PTB, respectively, were retrospectively analyzed from January 2017 to November 2022 in The Fourth Affiliated Hospital of Hebei Medical University (Ligang et al., A machine learning model based on CT and clinical features to distinguish pulmonary nodular mucinous adenocarcinoma from tuberculoma, 2023). A total of 1037 radiomic features were extracted from contrast-enhanced computed tomography (CT). The patients were randomly divided into a training group and a test group at a ratio of 7:3. The least absolute shrinkage and selection operator (LASSO) algorithm was used for radiomic feature selection. Three radiomics prediction models were applied: logistic regression (LR), support vector machine (SVM) and random forest (RF). The best performing model was adopted, and the radiomics score (Radscore) was then computed. The clinical model was developed using logistic regression. Finally, a combined model was established based on clinical factors and radiomics features. We externally validated the three models in a group of 68 patients (46 and 22 patients with PNMA and PTB, respectively) from Xing Tai People's Hospital (30 and 14 patients with PNMA and PTB, respectively) and The First Hospital of Xing Tai (16 and 8 patients with PNMA and PTB, respectively). The area under the receiver operating characteristic (ROC) curve (AUC) value and decision curve analysis were used to evaluate the predictive value of the developed models. RESULTS The combined model established by the logistic regression method had the best performance. The ROC-AUC (also a decision curve analysis) of the combined model was 0.940, 0.990 and 0.960 in the training group, test group and external validation group, respectively, and the combined model showed good predictive performance for the differentiation of PNMA from PTB. The Brier scores of the combined model were 0.132 and 0.068 in the training group and test group, respectively. CONCLUSION The combined model incorporating radiomics features and clinical parameters may have potential value for the preoperative differentiation of PNMA from PTB.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Computed Tomography and Magnetic Resonance, Hebei Medical University Fourth Affiliated Hospital, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Department of CT&MR, The First Hospital of Xing Tai, Xing Tai, 054000, He Bei, China
| | - Ligang Hao
- Department of Thoracic Surgery Xing, Tai People's Hospital, Xing Tai, 054000, He Bei, China
| | - MingWei Qi
- Department of Computed Tomography and Magnetic Resonance, Hebei Medical University Fourth Affiliated Hospital, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Qian Xu
- Department of Computed Tomography and Magnetic Resonance, Hebei Medical University Fourth Affiliated Hospital, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| | - Ning Zhang
- Department of Computed Tomography and Magnetic Resonance, Hebei Medical University Fourth Affiliated Hospital, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Hui Feng
- Department of Computed Tomography and Magnetic Resonance, Hebei Medical University Fourth Affiliated Hospital, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Gaofeng Shi
- Department of Computed Tomography and Magnetic Resonance, Hebei Medical University Fourth Affiliated Hospital, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
33
|
Zhuo Y, Li X, Feng W. NLRC3 is a potential prognostic biomarker that is correlated with immune cell infiltration in lung adenocarcinoma. Sci Rep 2023; 13:2923. [PMID: 36808166 PMCID: PMC9941471 DOI: 10.1038/s41598-022-23979-z] [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: 04/11/2022] [Accepted: 11/08/2022] [Indexed: 02/22/2023] Open
Abstract
The NLR family CARD domain containing 3 (NLRC3) gene has been reported to have a crucial effect on immunity, inflammation, and tumorigenesis. However, the clinical relevance of NLRC3 in lung adenocarcinoma (LUAD) remains unclear. This study analyzed both RNA sequencing data and corresponding clinical outcomes obtained from public databases to identify (i) NLRC3 as a tumor suppressor in LUAD and (ii) its predictive value for the likelihood of patient responsiveness to immunotherapy. The results showed that NLRC3 expression was reduced in LUAD and was lower in advanced-stage tumors. Additionally, reduced NLRC3 expression was correlated with worse patient prognosis. The protein level of NLRC3 was also observed to have prognostic significance. Moreover, downregulation of NLRC3 was found to suppress the chemotaxis and infiltration of antitumor lymphocyte subpopulations as well as natural killer cells. Mechanistic analysis indicated that NLRC3 may be involved in immune infiltration by regulating chemokines and their receptors in LUAD. Furthermore, NLRC3 functions as a molecular switch in macrophages, whereby it mediates the polarization of M1 macrophages. Patients with high NLRC3 expression were also found to exhibit a more promising response to immunotherapy. In conclusion, NLRC3 could serve as a potential prognostic biomarker for LUAD, help predict the immunotherapeutic response of patients, and guide personalized strategies for the treatment of LUAD.
Collapse
Affiliation(s)
- Yingchen Zhuo
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| | - Xueqian Li
- grid.452672.00000 0004 1757 5804Department of Endocrinology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710043 Shaanxi China
| | - Weiyi Feng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
34
|
Systemic Inflammation/Nutritional Status Scores Are Prognostic but Not Predictive in Metastatic Non-Small-Cell Lung Cancer Treated with First-Line Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24043618. [PMID: 36835030 PMCID: PMC9966997 DOI: 10.3390/ijms24043618] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Biomarkers of systemic inflammation/nutritional status have been associated with outcomes in advanced-stage non-small-cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs). However, most of them were not tested in cohorts of patients treated with ICIs in combination with chemotherapy (CT) (ICI + CT) or with CT alone, making it impossible to discriminate a predictive from a prognostic effect. We conducted a single-center retrospective study to search for associations between various baseline biomarkers/scores that reflected the systemic inflammation/nutritional status (Lung Immune Prognostic Index, Modified Lung Immune Prognostic Index, Scottish Inflammatory Prognostic Score, Advanced Lung Cancer Inflammation Index, EPSILoN, Prognostic Nutritional Index, Systemic Immune-Inflammation Index, Gustave Roussy Immune Score, Royal Marsden Hospital Prognostic Score, Lung Immuno-oncology Prognostic Score 3, Lung Immuno-oncology Prognostic Score 4, score published by Holtzman et al., and Glasgow Prognostic Score) and outcomes in metastatic NSCLC treated in a first-line setting either with ICI in monotherapy (cohort 1; n = 75), ICI + CT (cohort 2; n = 56), or CT alone (cohort 3; n = 221). In the three cohorts, the biomarkers/scores were moderately associated with overall survival (OS) and progression-free survival (PFS). Their prognostic performance was relatively poor, with a maximum c-index of 0.66. None of them was specific to ICIs and could help to choose the best treatment modality. The systemic inflammation/nutritional status, associated with outcomes independently of the treatment, is therefore prognostic but not predictive in metastatic NSCLC.
Collapse
|
35
|
In vivo induction of activin A-producing alveolar macrophages supports the progression of lung cell carcinoma. Nat Commun 2023; 14:143. [PMID: 36650150 PMCID: PMC9845242 DOI: 10.1038/s41467-022-35701-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
Alveolar macrophages (AMs) are crucial for maintaining normal lung function. They are abundant in lung cancer tissues, but their pathophysiological significance remains unknown. Here we show, using an orthotopic murine lung cancer model and human carcinoma samples, that AMs support cancer cell proliferation and thus contribute to unfavourable outcome. Inhibin beta A (INHBA) expression is upregulated in AMs under tumor-bearing conditions, leading to the secretion of activin A, a homodimer of INHBA. Accordingly, follistatin, an antagonist of activin A is able to inhibit lung cancer cell proliferation. Single-cell RNA sequence analysis identifies a characteristic subset of AMs specifically induced in the tumor environment that are abundant in INHBA, and distinct from INHBA-expressing AMs in normal lungs. Moreover, postnatal deletion of INHBA/activin A could limit tumor growth in experimental models. Collectively, our findings demonstrate the critical pathological role of activin A-producing AMs in tumorigenesis, and provides means to clearly distinguish them from their healthy counterparts.
Collapse
|
36
|
Jia H, Tang WJ, Sun L, Wan C, Zhou Y, Shen WZ. Pan-cancer analysis identifies proteasome 26S subunit, ATPase (PSMC) family genes, and related signatures associated with prognosis, immune profile, and therapeutic response in lung adenocarcinoma. Front Genet 2023; 13:1017866. [PMID: 36699466 PMCID: PMC9868736 DOI: 10.3389/fgene.2022.1017866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Background: Proteasome 26S subunit, ATPase gene (PSMC) family members play a critical role in regulating protein degradation and are essential for tumor development. However, little is known about the integrative function and prognostic significance of the PSMC gene family members in lung cancer. Methods: First, we assessed the expression and prognostic features of six PSMC family members in pan-cancer from The Cancer Genome Atlas (TCGA) dataset. Hence, by focusing on the relationship between PSMC genes and the prognostic, genomic, and tumor microenvironment features in lung adenocarcinoma (LUAD), a PSMC-based prognostic signature was established using consensus clustering and multiple machine learning algorithms, including the least absolute shrinkage and selection operator (LASSO) Cox regression, CoxBoost, and survival random forest analysis in TCGA and GSE72094. We then validated it in three independent cohorts from GEO and estimated the correlation between risk score and clinical features: genomic features (alterations, tumor mutation burden, and copy number variants), immune profiles (immune score, TIDE score, tumor-infiltrated immune cells, and immune checkpoints), sensitivity to chemotherapy (GDSC, GSE42127, and GSE14814), and immunotherapy (IMvigor210, GSE63557, and immunophenoscore). Twenty-one patients with LUAD were included in our local cohort, and tumor samples were submitted for evaluation of risk gene and PD-L1 expression. Results: Nearly all six PSMC genes were overexpressed in pan-cancer tumor tissues; however, in LUAD alone, they were all significantly correlated with overall survival. Notably, they all shared a positive association with increased TMB, TIDE score, expression of immune checkpoints (CD276 and PVR), and more M1 macrophages but decreased B-cell abundance. A PSMC-based prognostic signature was established based on five hub genes derived from the differential expression clusters of PSMC genes, and it was used to dichotomize LUAD patients into high- and low-risk groups according to the median risk score. The area under the curve (AUC) values for predicting survival at 1, 3, and 5 years in the training cohorts were all >.71, and the predictive accuracy was also robust and stable in the GSE72094, GSE31210, and GSE13213 datasets. The risk score was significantly correlated with advanced tumor, lymph node, and neoplasm disease stages as an independent risk factor for LUAD. Furthermore, the risk score shared a similar genomic and immune feature as PSMC genes, and high-risk tumors exhibited significant genomic and chromosomal instability, a higher TIDE score but lower immune score, and a decreased abundance of B and CD8+ T cells. Finally, high-risk patients were suggested to be less sensitive to immunotherapy but had a higher possibility of responding to platinum-based chemotherapy. The LUAD samples from the local cohort supported the difference in the expression levels of these five hub genes between tumor and normal tissues and the correlation between the risk score and PD-L1 expression. Conclusion: Overall, our results provide deep insight into PSMC genes in LUAD, especially the prognostic effect and related immune profile that may predict therapeutic responses.
Collapse
Affiliation(s)
- Hui Jia
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Wen-Jin Tang
- Department of Nursing, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Sun
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Chong Wan
- Yangtze Delta Region Institute of Tsinghua University, Jiaxing, China
| | - Yun Zhou
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Yun Zhou, ; Wei-Zhong Shen,
| | - Wei-Zhong Shen
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Yun Zhou, ; Wei-Zhong Shen,
| |
Collapse
|
37
|
Qiu W, Ren M, Wang C, Fu Y, Liu Y. The clinicopathological and prognostic significance of mTOR and p-mTOR expression in patients with non-small cell lung cancer: A meta-analysis. Medicine (Baltimore) 2022; 101:e32340. [PMID: 36595789 PMCID: PMC9794261 DOI: 10.1097/md.0000000000032340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/30/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) has a crucial role in carcinogenesis, angiogenesis, cellular proliferation, and metastasis; however, its significance in non-small cell lung cancer (NSCLC) remains contentious. Consequently, this study aims to assess the clinicopathological and prognostic importance of mTOR/p-mTOR expression in NSCLC. METHODS Literature retrieval was undertaken by searching English databases PubMed, EMBASE, Web of Science, and Cochrane Library as well as Chinese databases CNKI, Wan Fang, and VIP for full-text publications that satisfied our eligibility criteria up to November 2021. STATA 12.0 was used to conduct statistical analysis (STATA Corporation, College Station, TX). RESULTS This meta-analysis includes a total of 4683 patients from 28 primary publications. mTOR/p-mTOR expression was associated with sex (OR = 0.608, 95% CI: 0.442-0.836), lymph node metastasis (OR = 2.084, 95% CI: 1.437-3.182), and CEA (OR = 1.584, 95% CI: 1.135-2.209), but not with age, histological type, depth of tumor invasion, distant metastasis, TNM stage, differentiation degree, tumor size, or smoking. In addition, the expression of mTOR/p-mTOR is related to shorter overall survival in NSCLC patients (HR = 1.415, 95% CI: 1.051-1.905). CONCLUSION Positive mTOR/p-mTOR expression was substantially correlated with unfavorable conditions on the sex, lymph node metastases, and CEA levels. mTOR/p-mTOR may indicate a bad prognosis for NSCLC. The current findings must be confirmed and changed by other high-quality research employing a multivariate analysis on bigger sample size.
Collapse
Affiliation(s)
- Weiwei Qiu
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
- Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Meiying Ren
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Cuifeng Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yuhua Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
38
|
Tan S, Chen J, Wang Z, Zhao X, Sun Y, Zhu Y, Gong Z, Zhang D. Genomic characterization of thymic epithelial tumors reveals critical genes underlying tumorigenesis and poor prognosis. Clin Genet 2022; 103:529-539. [PMID: 36541162 DOI: 10.1111/cge.14285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Thymic epithelial tumors (TETs) are rare mediastinal tumors whose tumorigenesis mechanism is poorly understood. Characterization of molecular alterations in TETs may contribute to a better understanding of tumorigenesis and prognosis. Hybrid capture-based next-generation sequencing was performed on tumor tissues from 47 TETs (39 thymomas and 8 thymic carcinomas) to detect mutations in 315 tumor-associated genes. In total, 178 nonsynonymous mutations were identified, with a median of 3.79 per tumor in 47 TETs. Higher tumor mutation burden (TMB) level was more common in older TET patients, and significantly associated with the more advanced pathological type, especially in thymic carcinomas (TC) patients. The gene mutation profiles of B1-3, A/AB, and TC patients varied greatly. In the actionable mutations analysis, we found 32 actionable mutations in 24 genes. Among them, NFKBIA and TP53 mutations was the most frequently, which were only identified in TCs. Additionally, TCGA database analysis found that the expression of NFKBIA mRNA in the TCs were significantly higher than thymomas. TET patients with high NFKBIA expression had shorter overall survival compared with patients with low/medium NFKBIA expression, thus providing insights to consider NFKBIA as a potential prognosis biomarker and therapeutic target in TETs.
Collapse
Affiliation(s)
- Sheng Tan
- Department of Thoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jili Chen
- Department of Ophthalmology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zheng Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaokai Zhao
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing, China.,Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing, China
| | - Yonghua Sun
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing, China.,Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing, China
| | - Youcai Zhu
- Zhejiang Rongjun Hospital, Jiaxing, China
| | - Ziying Gong
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing, China.,Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing, China
| | - Daoyun Zhang
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing, China.,Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing, China
| |
Collapse
|
39
|
Kurose K, Sakaeda K, Fukuda M, Sakai Y, Yamaguchi H, Takemoto S, Shimizu K, Masuda T, Nakatomi K, Kawase S, Tanaka R, Suetsugu T, Mizuno K, Hasegawa T, Atarashi Y, Irino Y, Sato T, Inoue H, Hattori N, Kanda E, Nakata M, Mukae H, Oga T, Oka M. Immune checkpoint therapy and response biomarkers in non-small-cell lung cancer: Serum NY-ESO-1 and XAGE1 antibody as predictive and monitoring markers. Adv Clin Chem 2022; 112:155-204. [PMID: 36642483 DOI: 10.1016/bs.acc.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immune checkpoint inhibitors (ICI) are key drugs in systemic therapy for advanced non-small-cell lung cancer (NSCLC) and have recently been incorporated into neoadjuvant and adjuvant settings for surgical resection. Currently, ICI combinations with cytotoxic agents are frequently used in clinical practice, although several ICI clinical trials have failed to produce long-term clinical benefits. Unfortunately, clinical benefit is moderate and limited considering physical and financial burden. Therefore, selecting appropriate patients and regimens for ICI therapy is important, and biomarkers are necessary for their selection. Tumor PD-L1 expression is universally used as a biomarker; however, PD-L1 assays show low analytical validity and reproducibility due to the visual-scoring system by pathologists. Recent tumor immunology studies explore that neoantigens derived from somatic mutations and the collaboration between T and B cells efficiently elicit antitumor responses. This suggests that high tumor mutational burden and T-cell infiltration are predictive biomarkers. However, B cells producing antibody (Ab) remain poorly understood and analyzed as biomarkers. We found that NY-ESO-1 and XAGE1 of cancer-testis antigen frequently elicit spontaneous humoral and cellular immune responses in NSCLC. Serum Ab against these antigens were detected in approximately 25% of NSCLC patients and predicted ICI monotherapy responses. In addition, the Ab levels were decreased with tumor shrinkage after ICI therapy. Thus, NY-ESO-1 and XAGE1 Ab are potentially biomarkers predicting and monitoring response to ICI therapy. For clinical applications, a fully-automated assay system measuring the Ab was developed. Here, we review current ICI therapy, tumor immunology, and biomarkers in NSCLC, and discuss the applicability of the serum biomarkers NY-ESO-1 and XAGE1 Ab.
Collapse
Affiliation(s)
- Koji Kurose
- Department of Respiratory Medicine, Kawasaki Medical School, Okayama, Japan
| | - Kanako Sakaeda
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Minoru Fukuda
- Cancer Treatment Center, Nagasaki Prefecture Shimabara Hospital, Nagasaki, Japan
| | - Yumiko Sakai
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Hiroyuki Yamaguchi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shinnosuke Takemoto
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Takeshi Masuda
- Department of Respiratory Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Katsumi Nakatomi
- Department of Respiratory Medicine, NHO Ureshino Medical Center, Saga, Japan
| | - Shigeo Kawase
- Department of Respiratory Medicine, Kure Kyosai Hospital, Hiroshima, Japan
| | - Ryo Tanaka
- Department of Dermatology, Kawasaki Medical School, Okayama, Japan
| | - Takayuki Suetsugu
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Keiko Mizuno
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | | - Yusuke Atarashi
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Yasuhiro Irino
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Toshiyuki Sato
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima, Japan
| | - Eiichiro Kanda
- Department of Medical Science, Kawasaki Medical School, Okayama, Japan
| | - Masao Nakata
- General Thoracic Surgery, Kawasaki Medical School, Okayama, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toru Oga
- Department of Respiratory Medicine, Kawasaki Medical School, Okayama, Japan
| | - Mikio Oka
- Department of Immuno-Oncology, Kawasaki Medical School, Okayama, Japan.
| |
Collapse
|
40
|
Deng J, Xu W, Lei S, Li W, Li Q, Li K, Lyu J, Wang J, Wang Z. Activated Natural Killer Cells-Dependent Dendritic Cells Recruitment and Maturation by Responsive Nanogels for Targeting Pancreatic Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203114. [PMID: 36148846 DOI: 10.1002/smll.202203114] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/28/2022] [Indexed: 06/16/2023]
Abstract
Although enormous success has been obtained for dendritic cells (DCs)-mediated antigen-specific T cells anticancer immunotherapy in the clinic, it still faces major challenging problems: insufficient DCs in tumor tissue and low response rate for tumor cells lacking antigen expression, especially in low immunogenic tumors such as pancreatic cancer. Here, these challenges are tackled through tumor microenvironment responsive nanogels with prominent tumor-targeting capability by Panc02 cell membranes coating and inhibition of tumor-derived prostaglandin E2 (PGE2), aimed at improving natural killer (NK) cells activation and inducing activated NK cells-dependent DCs recruitment. The engineered nanogels can on-demand release acetaminophen to inhibit PGE2 secretion, thus promoting the activity of NK cells for non-antigen-specific tumor elimination. Furthermore, activated NK cells can secrete chemokines as CC motif chemokine ligand 5 and X-C motif chemokine ligand 1 to recruit immature DCs, and then promote DCs maturation and induce antigen-dependent CD8+ T cells proliferation for enhancing antigen-specific immunotherapy. Notably, these responsive nanogels show excellent therapeutic effect on Panc02 pancreatic tumor growth and postsurgical recurrence, especially combination of the programmed cell death-ligand 1 checkpoint-blockade immunotherapy. Therefore, this study provides a simple strategy for enhancing low immunogenic tumors immunotherapy through an antigen-independent way and antigen-dependent way synergetically.
Collapse
Affiliation(s)
- Junjie Deng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Weide Xu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Siyun Lei
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Wanyu Li
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qinghua Li
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Kaiqiang Li
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Jianxin Lyu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Zhen Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| |
Collapse
|
41
|
Noncoding RNAs-mediated overexpression of KIF14 is associated with tumor immune infiltration and unfavorable prognosis in lung adenocarcinoma. Aging (Albany NY) 2022; 14:8013-8031. [PMID: 36227151 PMCID: PMC9596199 DOI: 10.18632/aging.204332] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/23/2022] [Indexed: 11/28/2022]
Abstract
Kinesin family member 14 (KIF14) is potentially oncogenic and acts as a chromokinesin via binding to microtubules and chromatin during the bipolar spindle formation. KIF14 overexpression is a significant prognostic biomarker in various cancers. However, the expression, prognosis, mechanism, and tumor immune regulation of KIF14 in lung adenocarcinoma (LUAD) remain obscure. Our results demonstrated that KIF14 was upregulated in a variety of cancers, including LUAD. High-expression of KIF14 in LUAD was associated with pathological tumor stage, N stage and unfavorable prognosis. Both univariate and multivariate Cox regression results demonstrated that KIF14 was a significant independent risk factor influencing the prognosis of LUAD patients. The most promising upstream ncRNA-associated pathway of KIF14 in LUAD was determined to be GSEC/TYMSOS-hsa-miR-101-3p axis according to the starBase and The Cancer Genome Atlas databases. Furthermore, upregulation of KIF14 in LUAD was positively correlated with tumor mutation burden, microsatellite instability, immune checkpoint-related gene expression, immune cell biomarkers, and tumor immune cell infiltration. This study reveals that ncRNAs-mediated overexpression of KIF14 is associated with tumor immune infiltration and unfavorable prognosis in LUAD.
Collapse
|
42
|
Chen M, Jiang M, Wang X, Shen L, Li J. Envafolimab - first PD-1/PD-L1 antibody to be administered by subcutaneous injection for microsatellite instability-high or deficient mismatch repair advanced solid tumors. Expert Opin Biol Ther 2022; 22:1227-1232. [PMID: 36124972 DOI: 10.1080/14712598.2022.2125799] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors mobilize and activate the anti-tumor activity of the immune system by blocking the inhibitory effects of the PD-1/PD-L1 signaling pathway in T cells. Several anti-PD-1 or -PD-L1 monoclonal antibodies have been approved for the treatment of advanced solid tumors. However, most of immune checkpoint inhibitors (ICIs) are administered via intravenous infusion, which is inconvenient and leads to unsatisfactory patient compliance in the treatment process. Therefore, subcutaneous envafolimab is to be a potential treatment modality for advanced solid tumors. AREA COVERED A phase I clinical trial showed that the safety and pharmacokinetic profiles of envafolimab were similar to those of other traditional antibodies. Additionally, clinical findings from a phase II trial revealed that envafolimab monotherapy exhibited satisfactory clinical therapeutic effects and no significant adverse events in patients with Microsatellite instability-high/deficient mismatch Repair (MSI-H/dMMR) solid tumors who failed at least one line of prior systemic therapy. EXPERT OPINION Subcutaneous envafolimab may serve as a more convenient and acceptable treatment modality than those approved PD-1/PD-L1 inhibitors for patients with an advanced solid tumor, which may revolutionize the modes of immunotherapy in the future.
Collapse
Affiliation(s)
- Mifen Chen
- Department of Gastrointestinal Oncology, Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Mengyun Jiang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd. Nanjing, China
| | - Xinhui Wang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd. Nanjing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
43
|
Establishment of a Monoclonal Antibody-Based Enzyme-Linked Immunosorbent Assay to Measure Soluble B7-H5 in Patients with Cancer. J Immunol Res 2022; 2022:3013185. [PMID: 35966819 PMCID: PMC9371821 DOI: 10.1155/2022/3013185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
B7-H5, an immune checkpoint molecule, is markedly upregulated in multiple cancers and plays an important role in tumor progression and immune escape. However, the expression and significance of soluble B7-H5 (sB7-H5) in cancer remain unclear. Herein, we generated two novel mouse anti-human B7-H5 monoclonal antibodies (mAbs) 2E5 and 7B10, which had different epitopes. Based on the two mAbs, a sandwich enzyme-linked immunosorbent assay (ELISA) system was developed. Using this ELISA, we found that compared with healthy controls (HCs), sB7-H5 levels were significantly increased in the serum of patients with gastric cancer (GC), colorectal cancer (CRC), and lung cancer (LC) and were associated with TNM stage and metastasis. Receiver operating characteristic (ROC) curve analysis showed that sB7-H5 has diagnostic value for GC, CRC, and LC. Collectively, our findings delineate that sB7-H5 may be used as a predictor for diagnosis of cancer and a potential therapeutic target for cancer treatment.
Collapse
|
44
|
Prognosis of different extrathoracic metastasis patterns in patients with M1c lung adenocarcinoma receiving immunotherapy. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04182-z. [PMID: 35882652 DOI: 10.1007/s00432-022-04182-z] [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: 05/20/2022] [Accepted: 06/28/2022] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Lung cancer with extrathoracic metastases is classified as M1c. However, extrathoracic metastases can be further classified into different patterns. The purpose of this study was to analyze the survival differences between different patterns of extrathoracic metastases in patients with stage M1c lung adenocarcinoma after receiving immunotherapy. MATERIALS AND METHODS This study included 160 stage M1c lung adenocarcinoma patients and treated with immunotherapy. The enrolled patients were divided into two groups: those with multiple extrathoracic metastases alone (EM group) and those with simultaneous multiple extrathoracic and intrathoracic metastases (EIM group). Progression-free survival (PFS) and overall survival (OS) were evaluated. RESULTS The median PFS and OS in the whole group were 7.7 months and 25.4 months, respectively. The patients in the EM group show better PFS (13.0 months vs. 5.0 months; hazard ratio [HR] = 0.462, 95% confidence interval [CI] 0.317-0.673, P < 0.0001) and OS (35.0 months vs. 18.9 months; HR 0.592, 95% CI 0.380-0.922, P = 0.019) compared with the EIM group. Furthermore, in patients with lung adenocarcinoma with simultaneous extrathoracic and intrathoracic metastases who received immunotherapy, immunotherapy combined with chemotherapy has better PFS and OS than immunotherapy alone. There was no difference between immunotherapy alone or combined with chemotherapy in patients with lung adenocarcinoma with extrathoracic metastasis alone. CONCLUSION The different patterns of extrathoracic metastasis were related to the efficacy and prognosis of immunotherapy in M1c cohort. In addition, patients with simultaneous extrathoracic and intrathoracic metastases were more recommended to choose immunotherapy in combination with chemotherapy rather than immunotherapy alone.
Collapse
|
45
|
Thyroid Dysfunction as a Predictive Indicator in Camrelizumab of Advanced Esophageal Squamous Cell Carcinoma. J Immunol Res 2022; 2022:4015897. [PMID: 35832645 PMCID: PMC9273411 DOI: 10.1155/2022/4015897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022] Open
Abstract
Thyroid dysfunction (TD) induced by programmed death-1 (PD-1) or programmed cell death-ligand 1 (PD-L1) immune checkpoint inhibitors (ICIs) has been widely reported. However, the effects of ICI-induced TD on the survival of patients with esophageal squamous cell carcinoma (ESCC) have not been described. Herein, a retrospective study was conducted, which 82 patients with advanced metastatic or recurrent ESCC treated with camrelizumab were enrolled. Twenty patients (24.4%) experienced TD during camrelizumab treatment with or without chemotherapy. The median onset time of TD was 1.7 months. The incidence of TD was 35.6% in patients who previously received thoracic radiotherapy versus 10.8% in patients who did not (P =0.009). Patients with TD had significantly longer median progression-free survival (5.5 months vs 3.5 months, P =0.035) and overall survival (26.7 months vs 11.5 months, P <0.001). TD is frequently observed in ESCC patients treated with camrelizumab and especially in patients who received radiotherapy previously. ESCC patients with TD during ICIs treatment often have better prognosis.
Collapse
|
46
|
Zhu K, Yan A, Zhou F, Zhao S, Ning J, Yao L, Shang D, Chen L. A Pyroptosis-Related Signature Predicts Overall Survival and Immunotherapy Responses in Lung Adenocarcinoma. Front Genet 2022; 13:891301. [PMID: 35795208 PMCID: PMC9252528 DOI: 10.3389/fgene.2022.891301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Lung adenocarcinoma (LUAD) is a highly malignant cancer with a bleak prognosis. Pyroptosis is crucial in LUAD. The present study investigated the prognostic value of a pyroptosis-related signature in LUAD.Methods: LUAD’s genomic data were downloaded from TCGA and GEO databases. K-means clustering was used to classify the data based on pyroptosis-related genes (PRGs). The features of tumor microenvironment were compared between the two subtypes. Differentially expressed genes (DEGs) were identified between the two subtypes, and functional enrichment and module analysis were carried out. LASSO Cox regression was used to build a prognostic model. Its prognostic value was assessed.Results: In LUAD, genetic and transcriptional changes in PRGs were found. A total of 30 PRGs were found to be differentially expressed in LUAD tissues. Based on PRGs, LUAD patients were divided into two subgroups. Subtype 1 has a higher overall survival rate than subtype 2. The tumor microenvironment characteristics of the two subtypes differed significantly. Compared to subtype 1, subtype 2 had strong immunological infiltration. Between the two groups, 719 DEGs were discovered. WGCNA used these DEGs to build a co-expression network. The network modules were analyzed. A prognostic model based on seven genes was developed, including FOSL1, KRT6A, GPR133, TMPRSS2, PRDM16, SFTPB, and SFTA3. The developed model was linked to overall survival and response to immunotherapy in patients with LUAD.Conclusion: In LUAD, a pyroptosis-related signature was developed to predict overall survival and treatment responses to immunotherapy.
Collapse
Affiliation(s)
- Kaibin Zhu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - An Yan
- Department of Thoracic Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fucheng Zhou
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Su Zhao
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jinfeng Ning
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lei Yao
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Desi Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lantao Chen
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Lantao Chen,
| |
Collapse
|
47
|
Wang N, Yu M, Fu Y, Ma Z. Blocking ATM Attenuates SKOV3 Cell Proliferation and Migration by Disturbing OGT/OGA Expression via hsa-miR-542-5p. Front Oncol 2022; 12:839508. [PMID: 35795059 PMCID: PMC9251376 DOI: 10.3389/fonc.2022.839508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/25/2022] [Indexed: 12/13/2022] Open
Abstract
Blocking ataxia telangiectasia mutated (ATM), a crucial player in DNA repair responses, has been proposed as a promising strategy in anti-cancer therapy. Most previous studies have focused on DNA damage response-related pathways after administration of ATM inhibitors. However, ATM inhibition could potentially influence a wide range of changes in gene expression, which remain poorly defined. Here, we report that administration of the ATM inhibitor KU60019 led to impaired migration and enhanced apoptosis in the ovarian cancer cell line SKOV3, accompanied by abnormally elevated O-GlcNAc transferase and O-GlcNAcase expression levels. In addition, KU60019 treatment significantly suppressed expression of hsa-miR-542-5p in SKOV3 cells. Up-regulation of hsa-miR-542-5p expression inhibited increases in OGT and OGA level, and reversed the effects of ATM inhibition on apoptosis and migration in SKOV3 cells. Finally, we found aberrant expression of OGT and OGA to be associated with ovarian cancer patient survival. Taken together, our results suggest that ATM inhibition may promote SKOV3 cell apoptosis via suppressing hsa-miR-542-5p and elevating OGT and OGA expression, providing new insights into the application of ATM inhibitors in cancer immunotherapy.
Collapse
Affiliation(s)
- Ning Wang
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Department of Gynaecology II, The First Hospital of Jilin University, Changchun, China
| | - Miaomiao Yu
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Bethune Institute of Epigenetic Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yan Fu
- Department of Gynaecology I, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zhanchuan Ma, ; Yan Fu,
| | - Zhanchuan Ma
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zhanchuan Ma, ; Yan Fu,
| |
Collapse
|
48
|
Liu C, Liu D, Wang F, Xie J, Liu Y, Wang H, Rong J, Xie J, Wang J, Zeng R, Xie Y. The Interferon Gamma-Related Long Noncoding RNA Signature Predicts Prognosis and Indicates Immune Microenvironment Infiltration in Colon Adenocarcinoma. Front Oncol 2022; 12:876660. [PMID: 35747790 PMCID: PMC9211770 DOI: 10.3389/fonc.2022.876660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/16/2022] [Indexed: 12/17/2022] Open
Abstract
Colon adenocarcinoma (COAD) is one of the most common clinically malignant tumours of the digestive system, with high incidence and mortality and poor prognosis. Interferon-gamma (IFN-γ) and long noncoding RNAs (lncRNAs) have prognostic values and were closely associated with immune microenvironment in COAD. Thus, identifying IFN-γ-related lncRNAs may be valuable in predicting the survival of patients with COAD. In this study, we identified IFN-γ-related lncRNAs and divided COAD patients from the Cancer Genome Atlas (TCGA) database into training and validation sets. Pearson’s correlation analysis and least absolute shrinkage and selection operator (LASSO) Cox regression were performed to select IFN-γ-related lncRNA-associated prognoses. Thirteen lncRNAs (AC025165.8, AC091633.3, FENDRR, LINC00882, LINC01828, LINC01829, MYOSLID, RP11-154H23.4, RP11-20J15.3, RP11-324L17.1, RP11-342A23.2, RP11-805I24.3, SERTAD4-AS1) were identified to construct an IFN-γ-related lncRNA prognostic signature in TCGA training (n =213) and validation (n =213) cohorts. COAD patient risk scores were calculated and classified into high- and low-risk groups based on the median value of the risk scores in each dataset. We compared the overall survival (OS) of patients stratified by age, gender, and stage. The OS in the high-risk group was significantly shorter than that in the low-risk group. In addition, the clinical nomogram incorporating the prognostic signature and clinical features showed a high concordance index of 0.78 and accurately predicted 1-, 3-, and 5-year survival times among COAD patients in the high- and low-risk groups. Based on the risk model, the high- and low-risk groups exhibited distinct differences in the immune system by gene set enrichment analysis (GSEA) functional annotation, and differentially expressed genes (DEGs) between the high- and low-risk groups were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. We investigated the expression of multiple immune checkpoint genes in the high- and low-risk groups and plotted Kaplan-Meier survival curves, indicating that immune checkpoint genes, such as LAG3 and PD. L1, STING and TIM 3, were also expressed differently between the two risk groups. Subsequently, there were dramatic differences in mutated genes, SNV (single nucleotide variants) classes, variant types and variant allele frequencies between low- and high-risk patients with COAD. Patients stratified by risk scores had different sensitivities to common chemotherapeutic agents. Finally, we used quantitative real-time polymerase chain reaction (qRT-PCR) assays to demonstrate that three lncRNAs were significantly differentially expressed in COAD tissues and adjacent normal tissues. Considered together, a thirteen-lncRNA prognostic signature has great potential to be a prognostic biomarker and could play an essential role in the immune microenvironment of COAD.
Collapse
Affiliation(s)
- Cong Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Dingwei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Fangfei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Jun Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Yang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Huan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Jianfang Rong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Jinliang Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Jinyun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Rong Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
- *Correspondence: Yong Xie,
| |
Collapse
|
49
|
De Ridder K, Locy H, Piccioni E, Zuazo MI, Awad RM, Verhulst S, Van Bulck M, De Vlaeminck Y, Lecocq Q, Reijmen E, De Mey W, De Beck L, Ertveldt T, Pintelon I, Timmermans JP, Escors D, Keyaerts M, Breckpot K, Goyvaerts C. TNF-α-Secreting Lung Tumor-Infiltrated Monocytes Play a Pivotal Role During Anti-PD-L1 Immunotherapy. Front Immunol 2022; 13:811867. [PMID: 35493461 PMCID: PMC9046849 DOI: 10.3389/fimmu.2022.811867] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
Immune checkpoint blockade (ICB) of the PD-1 pathway revolutionized the survival forecast for advanced non-small cell lung cancer (NSCLC). Yet, the majority of PD-L1+ NSCLC patients are refractory to anti-PD-L1 therapy. Recent observations indicate a pivotal role for the PD-L1+ tumor-infiltrating myeloid cells in therapy failure. As the latter comprise a heterogenous population in the lung tumor microenvironment, we applied an orthotopic Lewis Lung Carcinoma (LLC) model to evaluate 11 different tumor-residing myeloid subsets in response to anti-PD-L1 therapy. While we observed significantly reduced fractions of tumor-infiltrating MHC-IIlow macrophages and monocytes, serological levels of TNF-α restored in lung tumor-bearing mice. Notably, we demonstrated in vivo and in vitro that anti-PD-L1 therapy mediated a monocyte-specific production of, and response to TNF-α, further accompanied by their significant upregulation of CD80, VISTA, LAG-3, SIRP-α and TIM-3. Nevertheless, co-blockade of PD-L1 and TNF-α did not reduce LLC tumor growth. A phenomenon that was partly explained by the observation that monocytes and TNF-α play a Janus-faced role in anti-PD-L1 therapy-mediated CTL stimulation. This was endorsed by the observation that monocytes appeared crucial to effectively boost T cell-mediated LLC killing in vitro upon combined PD-L1 with LAG-3 or SIRP-α blockade. Hence, this study enlightens the biomarker potential of lung tumor-infiltrated monocytes to define more effective ICB combination strategies.
Collapse
Affiliation(s)
- Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hanne Locy
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Elisa Piccioni
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Miren Ibarra Zuazo
- Immunomodulation Group, Navarrabiomed, Navarrabiomed-UPNA-IdISNA, Pamplona, Spain
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Mathias Van Bulck
- Laboratory of Molecular and Medical Oncology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Eva Reijmen
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Wout De Mey
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Lien De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology & Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology & Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| | - David Escors
- Immunomodulation Group, Navarrabiomed, Navarrabiomed-UPNA-IdISNA, Pamplona, Spain
- Rayne Institute, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
50
|
Liu C, Wang Z, Zeng Q, Zheng S, Feng X, Xue Q, Sun N, He J. PD-L1 expression and smoke exposure as biomarkers for optimizing adjuvant therapy for patients with resected limited-stage small-cell lung carcinoma. Genes Dis 2022; 10:48-50. [PMID: 37013059 PMCID: PMC10066250 DOI: 10.1016/j.gendis.2022.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Chengming Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhanyu Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qingpeng Zeng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Sufei Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Corresponding author. Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Corresponding author. Deparment of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|