1
|
WANG JIAHUI, GE HONGCHENG, YU ZHENGYUAN, WU LINGZHI. Non-coding RNAs as potential mediators of resistance to lung cancer immunotherapy and chemotherapy. Oncol Res 2025; 33:1033-1054. [PMID: 40296912 PMCID: PMC12034021 DOI: 10.32604/or.2024.058256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 04/30/2025] Open
Abstract
Lung cancer is a common cause of cancer-related death globally. The majority of lung cancer patients initially benefit from chemotherapy and immunotherapy. However, as the treatment cycle progresses and the disease evolves, the emergence of acquired resistance leads to treatment failure. Many researches have shown that non-coding RNAs (ncRNAs) not only influence lung cancer progression but also act as potential mediators of immunotherapy and chemotherapy resistance in lung cancer, mediating drug resistance by regulating multiple targets and pathways. In addition, the regulation of immune response by ncRNAs is dualistic, forming a microenvironment for inhibits/promotes immune escape through changes in the expression of immune checkpoints. The aim of this review is to understand the effects of ncRNAs on the occurrence and development of lung cancer, focusing on the role of ncRNAs in regulating drug resistance of lung cancer.
Collapse
Affiliation(s)
- JIAHUI WANG
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - HONGCHENG GE
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310018, China
| | - ZHENGYUAN YU
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - LINGZHI WU
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| |
Collapse
|
2
|
Figueroa A, Gholi SS, Jayant G, Wadgaonkar R, Gubran A, Kuperberg SJ. Improving diagnostic capabilities in lung cancer through next-generation sequencing: a narrative review. J Thorac Dis 2025; 17:476-486. [PMID: 39975740 PMCID: PMC11833572 DOI: 10.21037/jtd-24-488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/29/2024] [Indexed: 02/21/2025]
Abstract
Background and Objective Lung neoplasia is the leading cause of cancer death worldwide, thus, early detection and accuracy in establishing a diagnosis is paramount. As a consequence of decades of basic and translational studies revealing the genetic basis of lung cancer, a paradigm shift has occurred toward a personalized approach to medicine whereby mutational analysis confers an opportunity for safer, and expedient treatment options. In this context, next-generation sequencing (NGS) has emerged as a vital technological advance, and has become increasingly established as a core method for rapidly and effectively identifying actionable mutations in lung cancer. For these reasons, an updated review of the literature across invasive and non-invasive diagnostic modalities in lung cancer is warranted to inform diagnostic approaches and prompt new investigations. The objective of the present review is to provide a focused update on applications of NGS in lung cancer diagnostics, with a special focus on tissue acquisition methodologies and mutational analysis. Methods The search strategy included a survey of the current literature from 2005 to 2024 in PubMed, Medline, Scopus, and Google Scholar. Eligible study types included original research, literature reviews (narrative and systematic), and observational studies. which encompassed findings pertinent to the lung cancer diagnostics, mutational analysis and lung cancer treatment overlapping with applications and use of NGS technologies. Key Content and Findings There are extensive and diverse advantages to the use of NGS in lung cancer diagnostics, especially when compared to traditional sequencing techniques including, speed, effectiveness, easy adoption in the context of analysis of samples prepared for lung cancer diagnosis. Advances in cell-free DNA reinforce the firm role of NGS in novel approaches. Conclusions NGS implementation is a crucial and beneficial technological leap in lung cancer diagnosis, especially given the environment of novel and established targeted and immune based therapies which require mutational testing. Its numerous benefits such as expedient results and reduced sample requirements will continue to ability optimize lung cancer outcomes by virtue of improved patient safety, reduction of unnecessary procedures, and provision of accurate results.
Collapse
Affiliation(s)
- Andrew Figueroa
- Department of Pulmonary Medicine, Doylestown Hospital, Ambler, PA, USA
| | - Shadi Safar Gholi
- Department of Internal Medicine, New York City Health and Hospitals, New York, NY, USA
| | - Girish Jayant
- Department of Internal Medicine, Montefiore Medicine Center, The Bronx, NY, USA
| | - Raj Wadgaonkar
- College of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Ahmed Gubran
- Department of Internal Medicine, New York City Health and Hospitals, New York, NY, USA
| | - Stephen J. Kuperberg
- Department of Internal Medicine, New York City Health and Hospitals, New York, NY, USA
- New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
3
|
Kashyap P, Raj KV, Sharma J, Dutt N, Yadav P. Classification of NSCLC subtypes using lung microbiome from resected tissue based on machine learning methods. NPJ Syst Biol Appl 2025; 11:11. [PMID: 39824879 PMCID: PMC11742043 DOI: 10.1038/s41540-025-00491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025] Open
Abstract
Classification of adenocarcinoma (AC) and squamous cell carcinoma (SCC) poses significant challenges for cytopathologists, often necessitating clinical tests and biopsies that delay treatment initiation. To address this, we developed a machine learning-based approach utilizing resected lung-tissue microbiome of AC and SCC patients for subtype classification. Differentially enriched taxa were identified using LEfSe, revealing ten potential microbial markers. Linear discriminant analysis (LDA) was subsequently applied to enhance inter-class separability. Next, benchmarking was performed across six different supervised-classification algorithms viz. logistic-regression, naïve-bayes, random-forest, extreme-gradient-boost (XGBoost), k-nearest neighbor, and deep neural network. Noteworthy, XGBoost, with an accuracy of 76.25%, and AUROC (area-under-receiver-operating-characteristic) of 0.81 with 69% specificity and 76% sensitivity, outperform the other five classification algorithms using LDA-transformed features. Validation on an independent dataset confirmed its robustness with an AUROC of 0.71, with minimal false positives and negatives. This study is the first to classify AC and SCC subtypes using lung-tissue microbiome.
Collapse
Affiliation(s)
- Pragya Kashyap
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Kalbhavi Vadhi Raj
- Department of Electrical Engineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Jyoti Sharma
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Naveen Dutt
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Pankaj Yadav
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, Rajasthan, India.
| |
Collapse
|
4
|
Wang Y, Zhu Z, Luo R, Chen W. Single-cell transcriptome analysis reveals heterogeneity of neutrophils in non-small cell lung cancer. J Gene Med 2024; 26:e3690. [PMID: 38735760 DOI: 10.1002/jgm.3690] [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: 12/23/2023] [Revised: 03/23/2024] [Accepted: 04/07/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Lung cancer stands out as a highly perilous malignant tumor with severe implications for human health. There has been a growing interest in neutrophils as a result of their role in promoting cancer in recent years. Thus, the present study aimed to investigate the heterogeneity of neutrophils in non-small cell lung cancer (NSCLC). METHODS Single-cell RNA sequencing of tumor-associated neutrophils (TANs) and polymorphonuclear neutrophils sourced from the Gene Expression Omnibus database was analyzed. Moreover, cell-cell communication, differentiation trajectories and transcription factor analyses were performed. RESULTS Neutrophils were found to be closely associated with macrophages. Four major types of TANs were identified: a transitional subcluster that migrated from blood to tumor microenvironment (TAN-0), an inflammatory subcluster (TAN-1), a subpopulation that displayed a distinctive transcriptional signature (TAN-2) and a final differentiation state that promoted tumor formation (TAN-3). Meanwhile, TAN-3 displayed a marked increase in glycolytic activity. Finally, transcription factors were analyzed to uncover distinct TAN cluster-specific regulons. CONCLUSIONS The discovery of the dynamic characteristics of TANs in the present study is anticipated to contribute to yielding a better understanding of the tumor microenvironment and advancing the treatment of NSCLC.
Collapse
Affiliation(s)
- Yunzhen Wang
- 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
| | - Raojun Luo
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenwen Chen
- Nursing Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Su S, Chen F, Xu M, Liu B, Wang L. Recent advances in neoantigen vaccines for treating non-small cell lung cancer. Thorac Cancer 2023; 14:3361-3368. [PMID: 37905603 PMCID: PMC10693939 DOI: 10.1111/1759-7714.15126] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 11/02/2023] Open
Abstract
The breakthrough of programmed cell death protein 1 (PD-1) blockade therapy has changed the clinical treatment of non-small cell lung cancer (NSCLC) in the past few years. The success of PD-1 blockade therapy has been attributed to high tumor mutation burden and high immunogenicity of lung cancer cells. To further improve the efficacy of NSCLC immunotherapy and overcome the resistance of lung cancer cells to immune checkpoint blockade, new approaches that enhance the active immune response, such as neoantigen vaccines and cellular-based therapies, are urgently required. Neoantigens are considered ideal targets for cancer immunotherapy because of their high immunogenicity and specificity. In this mini review, we first discuss the current advances in neoantigen vaccines for treating cancers and then review the results of preclinical studies and early-phase human clinical trials of neoantigen-based therapies for NSCLC. Finally, we focus on the identification of neoantigens in patients with NSCLC and review the candidate mutations reported by recent studies and our investigations. The review concludes that, in addition to immune checkpoint blockade, approaches targeting neoantigens are promising for improving the efficacy of NSCLC immunotherapy.
Collapse
Affiliation(s)
- Shu Su
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjingChina
| | - Fungjun Chen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjingChina
| | - Mingyuan Xu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjingChina
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjingChina
| | - Lifeng Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjingChina
| |
Collapse
|
6
|
Xia Y, Gao Y, Liu MY, Li L, Pan W, Mao LZ, Yang Z, Yang M, Guo AY. ICBcomb: a comprehensive expression database for immune checkpoint blockade combination therapy. Brief Bioinform 2023; 25:bbad457. [PMID: 38095856 PMCID: PMC10753530 DOI: 10.1093/bib/bbad457] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/19/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
The success of immune checkpoint blockade (ICB) promotes the immunotherapy to be a new pillar in cancer treatment. However, the low response rate of the ICB therapy limits its application. To increase the response rate and enhance efficacy, the ICB combination therapy has emerged and its clinical trials are increasing. Nevertheless, the gene expression profile and its pattern of ICB combination were not comprehensively studied, which limits the understanding of the ICB combination therapy and the identification of new drugs. Here, we constructed ICBcomb (http://bioinfo.life.hust.edu.cn/ICBcomb/), a comprehensive database, by analyzing the human and mouse expression data of the ICB combination therapy and comparing them between groups treated with ICB, other drugs or their combinations. ICBcomb contains 1399 samples across 29 cancer types involving 52 drugs. It provides a user-friendly web interface for demonstrating the results of the available comparisons in the ICB combination therapy datasets with five functional modules: [1, 2] the 'Dataset/Disease' modules for browsing the expression, enrichment and comparison results in each dataset or disease; [3] the 'Gene' module for inputting a gene symbol and displaying its expression and comparison results across datasets/diseases; [4] the 'Gene Set' module for GSVA/GSEA enrichment analysis on the built-in gene sets and the user-input gene sets in different comparisons; [5] the 'Immune Cell' module for immune cell infiltration comparison between different groups by immune cell abundance analysis. The ICBcomb database provides the first resource for gene expression profile and comparison in ICB combination therapy, which may provide clues for discovering the mechanism of effective combination strategies and new combinatory drugs.
Collapse
Affiliation(s)
- Yun Xia
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yan Gao
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ming-Yu Liu
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lei Li
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wen Pan
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ling-Zi Mao
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhongzheng Yang
- Wuhan Biobank Co., Ltd., Wuhan Institute of Biotechnology, Wuhan, 430000, China
| | - Mei Yang
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - An-Yuan Guo
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Thoracic Surgery, West China Biomedical Big Data Center, Med-X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Teng Y, Wei Y, Chen Y, Yan J, Liu S, Li F, Bao S, Ren Y, Liu L, Yang Y. Patient preferences and shared decision making for the traditional Chinese medicine treatment of lung cancer: A discrete choice experiment study. Integr Med Res 2023; 12:100969. [PMID: 37546234 PMCID: PMC10400851 DOI: 10.1016/j.imr.2023.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 08/08/2023] Open
Abstract
Background Traditional Chinese Medicine (TCM), integrating patient preferences into decision-making process, has been widely used in the multimodality therapy of lung cancer. This study aimed to estimate patient preferences for treatment and shared decision-making (SDM) modes concerning inpatient TCM treatment of lung cancer in Shanghai in order to provide a basis for clinical decision-making process on TCM therapy for lung cancer. Methods This study was conducted among patients (n = 347) from nine tertiary hospitals in Shanghai by discrete-choice experiment (DCE) survey and Shared Decision-Making Questionnaire-patient version (SDM-Q-9) survey. The DCE was developed with the inclusion of the most relevant attributes at appropriate levels for the TCM treatment of lung cancer. The empirical data analyses of patients were performed using mixed logit models. Additionally, subgroup analysis was conducted. Results In total, 347 respondents completed the questionnaire. All attributes' coefficients were statistically significant for patients' preferences. Patients showed strong preferences for increasing disease control rate, relieving nausea and vomiting, reducing the risk of side effects, and were concerned about monthly out-of-pocket costs. Subgroup analysis indicated that patients with a lower SDM-Q-9 score and those who were satisfied with medical services emphasized more importance of higher disease control rate. Furthermore, most of the patients (90.20%) self-reported a high willingness to use SDM during the decision-making process. Conclusion In Shanghai, patients mainly preferred SDM and considered disease control rate as the most essential attribute in the TCM treatment of lung cancer. The study findings could underscore the importance of considering patients' preferences and promote SDM.
Collapse
Affiliation(s)
- Yue Teng
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Yan Wei
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Yingyao Chen
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Juntao Yan
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Shimeng Liu
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Fuming Li
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Shiyi Bao
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Yanfeng Ren
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Liu Liu
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Yi Yang
- School of Public Health, Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Khadela A, Postwala H, Rana D, Dave H, Ranch K, Boddu SHS. A review of recent advances in the novel therapeutic targets and immunotherapy for lung cancer. Med Oncol 2023; 40:152. [PMID: 37071269 DOI: 10.1007/s12032-023-02005-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/22/2023] [Indexed: 04/19/2023]
Abstract
Lung cancer is amongst the most pervasive malignancies having high mortality rates. It is broadly grouped into non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). The concept of personalized medicine has overshadowed the conventional chemotherapy given to all patients with lung cancer. The targeted therapy is given to a particular population having specific mutations to help in the better management of lung cancer. The targeting pathways for NSCLC include the epidermal growth factor receptor, vascular endothelial growth factor receptor, MET (Mesenchymal epithelial transition factor) oncogene, Kirsten rat sarcoma viral oncogene (KRAS), and anaplastic lymphoma kinase (ALK). SCLC targeting pathway includes Poly (ADP-ribose) polymerases (PARP) inhibitors, checkpoint kinase 1 (CHK 1) pathway, WEE1 pathway, Ataxia Telangiectasia and Rad3-related (ATR)/Ataxia telangiectasia mutated (ATM), and Delta-like canonical Notch ligand 3 (DLL-Immune checkpoint inhibitors like programmed cell death protein 1 (PD-1)/ programmed death-ligand 1 (PD-L1) inhibitors and Cytotoxic T-lymphocyte-associated antigen-4 (CTLA4) blockade are also utilized in the management of lung cancer. Many of the targeted therapies are still under development and require clinical trials to establish their safety and efficacy. This review summarizes the mechanism of molecular targets and immune-mediated targets, recently approved drugs, and their clinical trials for lung cancer.
Collapse
Affiliation(s)
- Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Humzah Postwala
- Pharm.D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Deval Rana
- Pharm.D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Hetvi Dave
- Pharm.D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Ketan Ranch
- Department of Pharmaceutics and Pharm. Technology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| |
Collapse
|
9
|
Tuli HS, Garg VK, Choudhary R, Iqubal A, Sak K, Saini AK, Saini RV, Vashishth K, Dhama K, Mohapatra RK, Gupta DS, Kaur G. Immunotherapeutics in lung cancers: from mechanistic insight to clinical implications and synergistic perspectives. Mol Biol Rep 2023; 50:2685-2700. [PMID: 36534236 DOI: 10.1007/s11033-022-08180-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lung cancer is one of the highly lethal forms of cancer whose incidence has worldwide rapidly increased over the past few decades. About 80-85% of all lung cancer cases constitute non-small cell lung cancer (NSCLC), with adenocarcinoma, squamous cell carcinoma and large cell carcinoma as the main subtypes. Immune checkpoint inhibitors have led to significant advances in the treatment of a variety of solid tumors, significantly improving cancer patient survival rates. METHODS AND RESULTS The cytotoxic drugs in combination with anti-PD-(L)1 antibodies is a new method that aims to reduce the activation of immunosuppressive and cancer cell prosurvival responses while also improving direct cancer cell death. The most commonly utilized immune checkpoint inhibitors for patients with non-small cell lung cancer are monoclonal antibodies (Atezolizumab, Cemiplimab, Ipilimumab, Pembrolizumab etc.) against PD-1, PD-L1, and CTLA-4. Among them, Atezolizumab (TECENTRIQ) and Cemiplimab (Libtayo) are engineered monoclonal anti programmed death ligand 1 (PD-L1) antibodies that inhibit binding of PD-L1 to PD-1 and B7.1. As a result, T-cell proliferation and cytokine synthesis are inhibited leading to restoring the immune homeostasis to fight cancer cells. CONCLUSIONS In this review article, the path leading to the introduction of immunotherapeutic options in lung cancer treatment is described, with analyzing the benefits and shortages of the current immunotherapeutic drugs. In addition, possibilities to co-administer immunotherapeutic agents with standard cancer treatment modalities are also considered.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana- Ambala, Haryana, 133 207, India.
| | - Vivek K Garg
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Gharuan, Mohali, Punjab, 140413, India
| | - Renuka Choudhary
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana- Ambala, Haryana, 133 207, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (Formerly, Faculty of Pharmacy), Jamia Hamdard (Deemed to Be University), Delhi, India
| | | | - Adesh K Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana- Ambala, Haryana, 133 207, India
| | - Reena V Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana- Ambala, Haryana, 133 207, India
| | - Kanupriya Vashishth
- Advance Cardiac Centre Department of Cardiology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, Odisha, 758002, India
| | - Dhruv Sanjay Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, 40056, India
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, 40056, India
| |
Collapse
|
10
|
Blichárová A, Tancoš V, Benetinová Z, Verbóová Ľ, Grendár M, Mazuráková A, Plank L, Mechírová E. Programmed death ligand-1 expression and its association with the degree of differentiation and the presence of necrosis in non-small cell lung carcinoma. Pathol Res Pract 2023; 242:154296. [PMID: 36610327 DOI: 10.1016/j.prp.2022.154296] [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: 11/18/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
The mechanisms underlying the expression of programmed death ligand-1 (PD-L1) in non-small cell lung carcinoma (NSCLC) are not yet fully clarified. In this study, surgical resections of 730 lung cancer patients with diagnosed NSCLC were analyzed. Results of PD-L1 immunohistochemistry (using clone 22C3) were correlated with clinicopathological variables including the degree of tumor differentiation and the presence of confluent areas of coagulative necrosis. PD-L1 immunohistochemistry was analyzed in tumor cells, whereas PD-L1 positivity was defined as membranous staining in ≥ 1 of tumor cells. A significantly higher proportion of PD-L1 positive cases was noted in poorly differentiated (grade 3) adenocarcinomas compared to better differentiated (grade 1 and grade 2) subtypes (63.8 % vs. 28.7 %; p < 0.001). Contrary to this, better differentiated (keratinizing) and less differentiated (non-keratinizing) squamous cell carcinoma subtypes were found to have a similar proportion of PD-L1 positive cases (51.4 % vs. 55.8 %; p = 0.570). High levels of PD-L1 expression significantly correlated with the presence of necrosis in NSCLC: seventy-nine of 109 NSCLC cases with the presence of necrosis were PD-L1 positive compared to 256 out of 621 NSCLC without necrosis (72.5 % vs. 41.2 %; p < 0.001). High PD-L1 expression was not positively correlated with age, gender, and advanced T stage but a significant association between PD-L1 positivity and higher N stage was observed (p < 0.001) in NSCLC patients. In conclusion, the proportion of PD-L1 positive cases is higher only in poorly differentiated NSCLC of the adenocarcinoma type. A significantly higher overall rate of PD-L1 positive cases was noted in NSCLC with the presence of necrosis. Further investigation is suggested to elucidate the intricated interconnections between the plethora of hypoxic biomarkers and immunological factors in different types and subtypes of NSCLC.
Collapse
Affiliation(s)
- Alžbeta Blichárová
- Department of Pathology, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Rastislavova 43, 040 01 Košice, Slovakia
| | - Vladimír Tancoš
- Department of Pathology, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Rastislavova 43, 040 01 Košice, Slovakia.
| | - Zuzana Benetinová
- Department of Pathology, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Rastislavova 43, 040 01 Košice, Slovakia
| | - Ľudmila Verbóová
- Department of Pathology, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Rastislavova 43, 040 01 Košice, Slovakia
| | - Marián Grendár
- Department of Bioinformatics, Biomedical Centre Martin, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Mala Hora 4C, 03601 Martin, Slovakia
| | - Alena Mazuráková
- Department of anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Kollárova 2, 03601 Martin, Slovakia
| | - Lukáš Plank
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Kollárova 2, 03659 Martin, Slovakia
| | - Eva Mechírová
- Department of Histology and Embryology, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Šrobárova 2, 041 80 Košice, Slovakia
| |
Collapse
|
11
|
Isla D, Lopez-Brea M, Espinosa M, Arrabal N, Pérez-Parente D, Carcedo D, Bernabé-Caro R. Cost-effectiveness of atezolizumab versus pembrolizumab as first-line treatment in PD-L1-positive advanced non-small-cell lung cancer in Spain. COST EFFECTIVENESS AND RESOURCE ALLOCATION 2023; 21:6. [PMID: 36647072 PMCID: PMC9841669 DOI: 10.1186/s12962-023-00417-z] [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: 08/04/2022] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Atezolizumab has recently been approved for first-line treatment of high PD-L1 expression metastatic Non-Small-Cell Lung Cancer (NSCLC) patients with no EGFR or ALK mutations, on the basis of the IMpower110 trial. This study aims to estimate the cost-effectiveness of atezolizumab compared with pembrolizumab among these patients in Spanish settings, based on the results of the two cut-offs of the IMpower110 study. METHODS A three-state partitioned-survival model was adapted to Spanish settings to calculate health outcomes and costs over a lifetime horizon. Clinical data for atezolizumab were collected from the interim and the exploratory results (data cut-off: Sept'18 and Feb'20, respectively) of the IMpower110 trial while a network meta-analysis was used to model pembrolizumab treatment. Utility data were collected from the trial. Direct medical costs were considered based on resources identified by experts. Costs and outcomes were discounted at 3% per year. Health outcomes were expressed as cost per Life Year (LY) and cost per Quality-Adjusted Life Year (QALY). Both deterministic and probabilistic sensitivity analyses were performed to assess the robustness of results. RESULTS Over a lifetime horizon, the incremental results showed that atezolizumab generated similar health outcomes (LYs and QALYs) to pembrolizumab, with minimal differences depending on the cut-off used (+ 0.70 and + 0.42 LYs and QALYs with Sept'18 cut-off and - 0.80 and - 0.72 LYs and QALYs with Feb'20 cut-off). However, for both cut-offs, atezolizumab produced meaningfully less costs than pembrolizumab (€ - 54,261 with Sept'18 cut-off and € - 81,907 with Feb'20 cut-off). The sensitivity analyses carried out confirmed the robustness of the base-case results. CONCLUSIONS The cost-effectiveness analysis, comparing the two cut-off of IMpower110, shows that atezolizumab provides similar health gains to pembrolizumab but at a lower cost for the first-line treatment of metastasic NSCLC patients in Spain.
Collapse
Affiliation(s)
- Dolores Isla
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | | | - María Espinosa
- Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | | | | | - Reyes Bernabé-Caro
- Hospital Universitario Virgen del Rocío, Sevilla, Spain
- Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
12
|
Amelia T, Setiawan AN, Kartasasmita RE, Ohwada T, Tjahjono DH. Computational Prediction of Resistance Induced Alanine-Mutation in ATP Site of Epidermal Growth Factor Receptor. Int J Mol Sci 2022; 23:ijms232415828. [PMID: 36555475 PMCID: PMC9784575 DOI: 10.3390/ijms232415828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) resistance to tyrosine kinase inhibitors can cause low survival rates in mutation-positive non-small cell lung cancer patients. It is necessary to predict new mutations in the development of more potent EGFR inhibitors since classical and rare mutations observed were known to affect the effectiveness of the therapy. Therefore, this research aimed to perform alanine mutagenesis scanning on ATP binding site residues without COSMIC data, followed by molecular dynamic simulations to determine their molecular interactions with ATP and erlotinib compared to wild-type complexes. Based on the result, eight mutations were found to cause changes in the binding energy of the ATP analogue to become more negative. These included G779A, Q791A, L792A, R841A, N842A, V843A, I853A, and D855A, which were predicted to enhance the affinity of ATP and reduce the binding ability of inhibitors with the same interaction site. Erlotinib showed more positive energy among G779A, Q791A, I853A, and D855A, due to their weaker binding energy than ATP. These four mutations could be anticipated in the development of the next inhibitor to overcome the incidence of resistance in lung cancer patients.
Collapse
Affiliation(s)
- Tasia Amelia
- School of Pharmacy, Bandung Institute of Technology, Jalan Ganesha 10, Bandung 40132, Indonesia
| | - Aderian Novito Setiawan
- School of Pharmacy, Bandung Institute of Technology, Jalan Ganesha 10, Bandung 40132, Indonesia
| | | | - Tomohiko Ohwada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daryono Hadi Tjahjono
- School of Pharmacy, Bandung Institute of Technology, Jalan Ganesha 10, Bandung 40132, Indonesia
- Correspondence: ; Tel.: +62-22-250-4852
| |
Collapse
|
13
|
Freire T, Landeira M, Giacomini C, Festari MF, Pittini Á, Cardozo V, Brosque A, Monin L, da Costa V, Faral-Tello P, Robello C, Osinaga E. Trypanosoma cruzi-Derived Molecules Induce Anti-Tumour Protection by Favouring Both Innate and Adaptive Immune Responses. Int J Mol Sci 2022; 23:ijms232315032. [PMID: 36499361 PMCID: PMC9739173 DOI: 10.3390/ijms232315032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
Lung cancer remains the leading cause of cancer mortality worldwide. Thus, the development of strategies against this type of cancer is of high value. Parasite infections can correlate with lower cancer incidence in humans and their use as vaccines has been recently explored in preclinical models. In this study, we investigated whether immunisations with a Trypanosoma cruzi lysate from epimastigotes protect from lung tumour growth in mice. We also explore the role of parasite glycans in the induction of the protective immune response. A pre-clinical murine cancer model using the lung tumour cell line LL/2 was used to evaluate the anti-tumour potential, both in preventive and therapeutic settings, of a T. cruzi epimastigote-derived protein lysate. Immunisation with the parasite lysate prevents tumour growth and induces both humoral and cellular anti-tumour immune responses to LL-2 cancer cells. The induced immunity and tumour protection were associated with the activation of natural killer (NK) cells, the production of interferon-γ (IFN-γ) and tumour cell cytotoxicity. We also show that mannose residues in the T. cruzi lysate induce Toll-like receptor (TLR) signalling. The evaluated T. cruzi lysate possesses anti-tumour properties likely by activating innate and adaptive immunity in a process where carbohydrates seem to be essential.
Collapse
Affiliation(s)
- Teresa Freire
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
- Correspondence: (T.F.); (E.O.)
| | - Mercedes Landeira
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Cecilia Giacomini
- Laboratorio de Bioquímica, Departamento de Biociencias, Facultad de Química, UdelaR, Gral Flores 2124, Montevideo 11800, Uruguay
| | - María Florencia Festari
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Álvaro Pittini
- Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Viviana Cardozo
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Alina Brosque
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Leticia Monin
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Valeria da Costa
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Paula Faral-Tello
- Laboratorio de Interacciones Hospedero-Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero-Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Eduardo Osinaga
- Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Correspondence: (T.F.); (E.O.)
| |
Collapse
|
14
|
Peng C, Chen J, Cui W, Li S, Li J, Peng L. Comparative efficacy of various CHIs combined with western medicine for non-small cell lung cancer: A bayesian network meta-analysis of randomized controlled trials. Front Pharmacol 2022; 13:1037620. [PMID: 36438813 PMCID: PMC9686447 DOI: 10.3389/fphar.2022.1037620] [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: 09/06/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
Background: Given the limitations of Western medicine (WM) for the treatment of non-small cell lung cancer (NSCLC) and the wide exploration of Chinese herbal injections (CHIs), systematically evaluate the efficacy of Various CHIs Combined with WM for Non-small Cell Lung Cancer. In this study, we performed a network meta-analysis to evaluate the comparative efficacy of 16 CHIs combined with WM regimens for the treatment of NSCLC. Methods: Literature databases were searched from their inception to November 2021, and all randomized control trials (RCTs) involving NSCLC patients treated with a combination of Chinese and WM were retrieved. Outcomes, including disease control rate, survival quality score, incidence of gastrointestinal adverse reactions, incidence of leukopenia, and incidence of thrombocytopenia, were analyzed using RevMan (5.3), Stata17, and R software. Surface under the cumulative ranking curve (SUCRA) probability values were calculated to rank the treatments examined, and clustering analysis was used to compare the effects of CHIs on different outcomes. Results: A total of 389 studies involving 31,263 patients and 16 CHIs were included. The 16 CHIs were: Aidi injection (ADI), Huachansu injection (HCSI), oil of Ophiopogon injection (OOMI), disodium cantharidinate and vitamin B6 injection (DCI), Shenfu injection (SFI), Shenmai injection (SMI), Shenqi Fuzheng injection (SQFZI), Chansu injection (CSI), Delisheng injection (DLSI), Fufang Kushen injection (FFKSI), Huangqi injection (HQI), Kangai injection (KAI), Kanglaite injection (KLTI), Shengmai injection (SI), Xiangguduotang injection (XGDTI), and Xiaoaiping injection (XAPI). The results of the network meta-analysis showed that, with WM treatment as a co-intervention, CSI was most likely to improve the disease control rate (SUCRA = 80.90%), HQI had the highest probability of being the best option for improving the survival quality score (SUCRA = 82.60%), DCI had the highest probability of reducing the incidence of gastrointestinal adverse reactions (SUCRA = 85.50%), HCSI + WM had the highest probability of reducing the incidence of thrombocytopenia (SUCRA = 91.30%), while SMI had the highest probability of reducing the incidence of leukopenia (SUCRA = 79.10%). Conclusion: CHIs combined with WM is proved to be more effective than WM alone, which may be beneficial to NSCLC patients. SMI + WM and DCI + WM are most likely the optimal CHI to improve disease control rates, survival quality score, and reduce adverse effects. This study has limitations; therefore, higher quality RCTs and real-world evidence are required to support our conclusions.
Collapse
Affiliation(s)
- Ciyan Peng
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Chen
- Department of Pharmacy, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Wei Cui
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Sini Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jianhe Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liubao Peng
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Liubao Peng,
| |
Collapse
|
15
|
Pal K, Hussain T, Xie H, Li S, Yang P, Mansfield A, Lou Y, Chowdhury S, Mukhopadhyay D. Expression, correlation, and prognostic significance of different nicotinic acetylcholine receptors, programed death ligand 1, and dopamine receptor D2 in lung adenocarcinoma. Front Oncol 2022; 12:959500. [PMID: 36072788 PMCID: PMC9441878 DOI: 10.3389/fonc.2022.959500] [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: 06/01/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Objective The objective of this study is to evaluate the expression of different nicotinic acetylcholine receptors (nAChRs), programmed death ligand-1 (PD-L1), and dopamine receptor D2 (DRD2) as prognostic factors in lung cancer and any correlation among them. Since all of the above genes are typically upregulated in response to smoking, we hypothesized that a correlation might exist between DRD2, PD-L1, and nAChR expression in NSCLC patients with a smoking history and a prediction model may be developed to assess the clinical outcome. Methods We retrospectively analyzed samples from 46 patients with primary lung adenocarcinoma who underwent surgical resection at Mayo Clinic Rochester from June 2000 to October 2008. The expression of PD-L1, DRD2, CHRNA5, CHRNA7, and CHRNA9 were analyzed by quantitative PCR and correlated amongst themselves and with age, stage and grade, smoking status, overall survival (OS), and relapse-free survival (RFS). Results Only PD-L1 showed a statistically significant increase in expression in patients older than 65. All the above genes showed higher expression in stage IIIB than IIIA, but none reached statistical significance. Interestingly, we did not observe significant differences among never, former, and current smokers, but patients with pack years greater than 30 showed significantly higher expression of CHRNA9. We observed a strong positive correlation between PD-L1/DRD2, PD-L1/CHRNA5, and CHRNA5/CHRNA7 and a weak positive correlation between DRD2/CHRNA5 and DRD2/CHRNA7. Older age was independently associated with poor OS, whereas lower CHRNA7 expression was independently associated with better OS. Conclusions We observed strong positive correlations among PD-L1, DRD2, and some of the nAChRs. We investigated their prognostic significance in lung cancer patients and found CHRNA7 to be an independent prognostic factor. Overall, the results obtained from this preliminary study warrant a large cohort-based analysis that may ultimately lead to potential patient-specific stratification biomarkers predicting cancer-treatment outcomes.
Collapse
Affiliation(s)
- Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Tabish Hussain
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Hao Xie
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Shenduo Li
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Ping Yang
- Department of Quantitative Health Sciences, Mayo Clinic Scottsdale, AZ, United States
| | - Aaron Mansfield
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Yanyan Lou
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Shantanu Chowdhury
- Integrative and Functional Biology Unit, CSIR- Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR- Institute of Genomics and Integrative Biology, New Delhi, India
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
16
|
Li X, Zhou X, Liu J, Zhang J, Feng Y, Wang F, He Y, Wan A, Filipczak N, Yalamarty SSK, Jin Y, Torchilin VP. Liposomal Co-delivery of PD-L1 siRNA/Anemoside B4 for Enhanced Combinational Immunotherapeutic Effect. ACS APPLIED MATERIALS & INTERFACES 2022; 14:28439-28454. [PMID: 35726706 DOI: 10.1021/acsami.2c01123] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Combination therapy has gained a lot of attention thanks to its superior activity against cancer. In the present study, we report a cRGD-targeted liposomal preparation for co-delivery of programmed cell death ligand 1 (PD-L1) small interfering RNA (siRNA) and anemoside B4 (AB4)─AB4/siP-c-L─and evaluate its anticancer efficiency in mouse models of LLC and 4T1 tumors. AB4/siP-c-L showed a particle size of (180.7 ± 7.3) nm and a ζ-potential of (32.8 ± 1.5) mV, with high drug encapsulation, pH-sensitive release properties, and good stability in serum. AB4/siP-c-L demonstrated prolonged blood circulation and increased tumor accumulation. Elevated cellular uptake was dependent on the targeting ligand cRGD. This combination induced significant tumor inhibition in LLC xenograft tumor-bearing mice by downregulating PD-L1 protein expression and modulating the immunosuppressive microenvironment. Liposomes favored the antitumor T-cell response with long-term memory, without obvious toxicity. A similar tumor growth inhibition was also demonstrated in the 4T1 tumor model. In summary, our results indicate that cRGD-modified and AB4- and PD-L1 siRNA-coloaded liposomes have potential as an antitumor preparation, and this approach may lay a foundation for the development of a new targeted drug delivery system.
Collapse
Affiliation(s)
- Xiang Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiong Zhou
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jun Liu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yulin Feng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Fang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yao He
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Anping Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Satya Siva Kishan Yalamarty
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
17
|
Deng J, Lin X, Li Q, Cai XY, Wu LW, Wang W, Zhang B, Li YL, Hu J, Lin NM. Decreased INPP5B expression predicts poor prognosis in lung adenocarcinoma. Cancer Cell Int 2022; 22:189. [PMID: 35568951 PMCID: PMC9107680 DOI: 10.1186/s12935-022-02609-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Inositol Polyphosphate-5-Phosphatase B (INPP5B), a inositol 5-phosphatase, plays an important role in many biological processes through phosphorylating PI(4,5)P2 and/or PI(3,4,5)P3 at the 5-position. Nevertheless, little is known about its function and cellular pathways in tumors. This study aims to investigate the potential role of INPP5B as a diagnostic and prognostic biomarker for lung adenocarcinoma (LUAD), as well as its biological functions and molecular mechanisms in LUAD. Methods TCGA, GEO, CTPAC, and HPA datasets were used for differential expression analysis and pathological stratification comparison. The prognostic and diagnostic role of INPP5B was determined by Kaplan–Meier curves, univariate and multivariate Cox regression analysis, and receiver operating characteristics (ROC) curve analyses. The potential mechanism of INPP5B was explored through GO, KEGG, and GSEA enrichment analysis, as well as GeneMANIA and STRING protein–protein interaction (PPI) network. PicTar, PITA, and miRmap databases were used for exploring miRNA targeting INPP5B. In molecular biology experiments, immunohistochemical analyses and Western blot analyses were used to determine protein expression. Co-immunoprecipitation assay was used to detect protein–protein interactions. CCK8 assays and colony formation assays were used for the measurement of cell proliferation. Cell cycle was assessed by PI staining with flow cytometry. Cell migration was performed by Transwell assays and wound healing assays. Result INPP5B was decreased in LUAD tissues compared with normal adjacent tissues. And the low expression of INPP5B was associated with late-stage pathological features. In addition, INPP5B was found to be a significant independent prognostic and diagnostic factor for LUAD patients. Hsa-miR-582-5p was predicted as a negative regulator of INPP5B mRNA expression. INPP5B was significantly correlated with the expression of PTEN and the activity of PI3K/AKT signaling pathways, as determined by enrichment analysis and PPI network. In vitro experiments partially confirmed the aforementioned findings. INPP5B could interact directly with PTEN. INPP5B overexpression inhibited LUAD cell proliferation and migration while downregulating the AKT pathway. Conclusion Our results demonstrated that INPP5B could inhibit the proliferation and metastasis of LUAD cells. It could serve as a novel diagnostic and prognostic biomarker for LUAD patients. Trial registration LUAD tissues and corresponding para-cancerous tissues were collected from 10 different LUAD patients at Hangzhou First People’s Hospital. The Ethics Committee of Hangzhou First People’s Hospital has approved this study. (registration number: IIT-20210907-0031-01; registration date: 2021.09.13) Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02609-8.
Collapse
Affiliation(s)
- Jun Deng
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qi Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiao-Yu Cai
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lin-Wen Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wei Wang
- Department of Pathology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yang-Ling Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Neng-Ming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China. .,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China. .,Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Lee H, Kwon YJ, Jin H, Liu H, Kang W, Chun YJ, Bae J, Choi HK. Anticancer activity and metabolic profile alterations by ortho-topolin riboside in in vitro and in vivo models of non-small cell lung cancer. FASEB J 2022; 36:e22127. [PMID: 35066937 DOI: 10.1096/fj.202101333r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2024]
Abstract
Lung cancer has the highest incidence and mortality rates among all types of cancer worldwide, and 80%-85% of patients with lung cancer are diagnosed with non-small cell lung cancer (NSCLC), which has 5-year survival rate of only 5% at advanced stages. Development of new therapeutic agents and strategies is required to enhance the treatment efficiency in patients with NSCLC. Metabolic alterations and anticancer effects of plant hormones and their derivatives have not been investigated in NSCLC in vitro and in vivo. The present study investigated the cytotoxic effects of 11 plant hormones and their derivatives against NSCLC cell lines; ortho-topolin riboside (oTR) showed the highest cytotoxicity among all tested compounds against NSCLC cells. Alteration of metabolites and lipids was investigated using gas chromatography-mass spectrometry and nano electrospray ionization-mass spectrometry in oTR-treated NSCLC cells and a xenograft mouse model. oTR reduced amino acid and pyrimidine synthesis in NSCLC cells and xenograft tumors. Moreover, oTR reduced glycolytic function and decreased mitochondrial respiration function by inhibiting glutamine and fatty acid oxidation. Increased levels of phosphatidylcholine, phosphatidylethanolamine, and phosphatidylserine species suggested that oTR might act as a fatty acid oxidation inhibitor. In addition, the increased level of phosphatidylserine species implied that phosphatidylserine-mediated apoptosis occurred in oTR-treated NSCLC cells and xenograft tumor. The antiproliferative and apoptotic effects of oTR were mediated by the reduced p-ERK and p-AKT levels and increased cleaved Caspase-3 levels, respectively. This is the first study to investigate the metabolic alterations and anticancer activity of oTR in in vitro and in vivo models of NSCLC. Our results provide basis for the development of oTR-based therapeutic agent for patients with NSCLC.
Collapse
Affiliation(s)
- Hwanhui Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hanyong Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Heifeng Liu
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Wonku Kang
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Jeehyeon Bae
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Tu J, Xu H, Ma L, Li C, Qin W, Chen X, Yi M, Sun L, Liu B, Yuan X. Nintedanib enhances the efficacy of PD-L1 blockade by upregulating MHC-I and PD-L1 expression in tumor cells. Theranostics 2022; 12:747-766. [PMID: 34976211 PMCID: PMC8692903 DOI: 10.7150/thno.65828] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs), such as programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1), have been widely applied in clinical and scientific research. Despite their effective antitumor effects in clinical tumor therapy, most tumors are still resistant to ICIs and long-term benefits are lacking. In addition, tumor patients complicated with interstitial lung disease limit the application of ICI therapy. Therefore, for these cases, there is an urgent need to develop new methods to relieve lung complications and enhance the efficacy of ICI therapy. Nintedanib, a potent triple angiokinase inhibitor approved for the treatment of progressive fibrotic interstitial lung disease. However, its immunotherapy synergy properties and mechanism are still pending further exploration. Methods: To explore the therapeutic potential of nintedanib and αPD-L1 combination therapy, MC38, LLC, and 4T1 tumor models were used to investigate antitumor and antimetastatic activities in vivo. An idiopathic pulmonary fibrosis-tumor bearing model was used to evaluate the effect of the synergy therapy on tumor model complicated with lung disease. Moreover, RNA-seq, immunohistochemistry, and flow cytometry were utilized to analyze the effect of combination treatment on the tumor microenvironment. The bioactivity following different treatments was determined by western blotting, CCK-8, and flow cytometry. Results: In this study, nintedanib and αPD-L1 synergy therapy exhibited significant antitumor, antimetastatic and anti-pulmonary fibrosis effects. Both in vitro and in vivo experiments revealed that these effects included promoting vessel normalization, increasing infiltration and activation of immune cells in tumors, enhancing the response of interferon-gamma, and activating the MHC class I-mediated antigen presentation process. Moreover, our results showed an increased expression of PD-L1 and promoted phosphorylation of STAT3 after nintedanib (1 µM) treatment. Conclusion: The combination of nintedanib and αPD-L1 increased ICI therapy responses, relieved lung complications and further activated the tumor immune microenvironment; thus, exhibiting a notable antitumor effect. Accordingly, the nintedanib synergy strategy is expected to be a promising candidate therapy for tumor patients complicated with interstitial lung disease in clinical practice.
Collapse
|
20
|
Yan J, Wei Y, Teng Y, Liu S, Li F, Bao S, Ren Y, Chen Y. Physician Preferences and Shared-Decision Making for the Traditional Chinese Medicine Treatment of Lung Cancer: A Discrete-Choice Experiment Study in China. Patient Prefer Adherence 2022; 16:1487-1497. [PMID: 35747587 PMCID: PMC9211799 DOI: 10.2147/ppa.s365109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/04/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND With progress being made in the treatment of cancer, various clinical and treatment options are being pursued. In China, Traditional Chinese Medicine (TCM) is used widely in the treatment of cancer. OBJECTIVE To estimate TCM treatment preferences and SDM mode of physicians in China. METHODS This study was conducted among physicians (n=185) from nine tertiary hospitals in China by discrete-choice experiment (DCE) survey and Shared Decision-Making Questionnaire-physician version (SDM-Q-Doc) survey. The DCE was developed with the inclusion of the most relevant attributes at appropriate levels for the TCM treatment of lung cancer. The empirical data analyses of physicians were performed using mixed logit models. Additionally, subgroup analysis was conducted. RESULTS In total, 185 respondents completed the questionnaire. All attributes were statistically significant except out-of-pocket costs. Physicians showed the strongest preferences for increasing disease control rate, relieving nausea and vomiting, and reducing the risk of side effects. Most of the physicians (78.38%) self-reported a high willingness to use SDM during the decision-making process. The physicians with a higher SDM-Q-Doc score had more preference for improving all three attributes than those with a lower score. Little variation was found in preferences among the physicians with other sociodemographic characteristics. CONCLUSION In China, physicians considered disease control rate as the most essential attribute in the TCM treatment of lung cancer. The physicians in China mainly preferred SDM, and the preference was different according to SDM mode when involving the TCM therapy for patients with lung cancer. The study findings could inform future TCM therapy for lung cancer and promote SDM.
Collapse
Affiliation(s)
- Juntao Yan
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
| | - Yan Wei
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
- Correspondence: Yan Wei, National Health Commission Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai, People’s Republic of China, Tel +86-18930749707, Email
| | - Yue Teng
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
- Outpatient Department of Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Shimeng Liu
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
| | - Fuming Li
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
| | - Shiyi Bao
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
| | - Yanfeng Ren
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
| | - Yingyao Chen
- School of Public Health, Fudan University, Shanghai, People’s Republic of China
- National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
21
|
Nierstedt RT, Yeahia R, Barnett KM. Unanticipated Myocarditis in a Surgical Patient Treated With Pembrolizumab: A Case Report. A A Pract 2021; 14:e01177. [PMID: 32132361 DOI: 10.1213/xaa.0000000000001177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We report a case of fatal immune checkpoint inhibitor (ICI)-associated myocarditis in a 77-year-old man with metastatic non-small cell lung cancer (NSCLC) who presented for mediport placement at our outpatient surgical center. He denied any cardiac complaints and had a previously normal electrocardiogram (EKG) off treatment. Intraoperatively and postoperatively, he displayed cardiac rhythm abnormalities. The patient was then transferred to a tertiary facility, where he expired within 48 hours. As cancer immunotherapy becomes increasingly prominent, ICI-associated myocarditis should be considered a potentially critical contributor to perioperative cardiac morbidity and mortality.
Collapse
Affiliation(s)
- Ryan T Nierstedt
- From the Oakland University William Beaumont School of Medicine, Rochester, Michigan.,Department of Anesthesiology and Critical Care, Memorial Sloan Kettering Cancer Center Monmouth Ambulatory Surgery Center, Middletown, New Jersey
| | - Rubaya Yeahia
- Department of Anesthesiology and Critical Care, Memorial Sloan Kettering Cancer Center Monmouth Ambulatory Surgery Center, Middletown, New Jersey.,New York Medical College, Valhalla, New York
| | - Kara M Barnett
- Department of Anesthesiology and Critical Care, Memorial Sloan Kettering Cancer Center Monmouth Ambulatory Surgery Center, Middletown, New Jersey
| |
Collapse
|
22
|
Huang M, Li T, Wang Q, Li C, Zhou H, Deng S, Lv Z, He Y, Hou B, Zhu G. Silencing circPVT1 enhances radiosensitivity in non-small cell lung cancer by sponging microRNA-1208. Cancer Biomark 2021; 31:263-279. [PMID: 33896835 DOI: 10.3233/cbm-203252] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Radiotherapy is one of main useful therapies in non-small cell lung cancer (NSCLC). Nevertheless, the underlying mechanism between NSCLC cell radiosensitivity and effective treatment remains unclear. OBJECTIVE The aim is to explore the relationship between circular (circ) RNA and NSCLC cell radiosensitivity. METHODS CircRNA plasmacytoma variant translocation 1 (PVT1) and microRNA (miR)-1208 expression in NSCLC cells were assessed using quantitative reverse transcriptase PCR (qRT-PCR). NSCLC cells were transfected with si-PVT1 or miR-1208 inhibitor and then exposed to irradiation. Cellular biology behaviors were detected using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL), colony formation, invasion and western blot. Additionally, binding between circPVT1 and miR-1208 was testified by dual-luciferase reporter and RIP assay. RESULTS CircPVT1 was upregulated in NSCLC cells after irradiation treatment. Silencing circPVT1 induced inhibition of NSCLC cell growth and invasion, accompanied by cell apoptosis and γ-H2AX expression. Moreover, NSCLC cell proliferation and invasion was further inhibited by irradiation treatment in circPVT1-silenced cells, indicating a strong radiosensitivity of NSCLC cells. CircPVT1 functions as a competing endogenous RNA of miR-1208. Silencing miR-1208 reversed NSCLC cell sensitivity response to irradiation and activated PI3K/AKT/mTOR pathway in circPVT1-silenced cells. CONCLUSIONS Silencing circPVT1 enhanced radiosensitivity of NSCLC cells by sponging miR-1208.
Collapse
Affiliation(s)
- Meifang Huang
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China.,Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Tianqian Li
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China.,Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Qing Wang
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Chongxin Li
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Huahua Zhou
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Shengyi Deng
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Zengbo Lv
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Yongmei He
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Bo Hou
- Department of Thoracic Surgery, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Guangying Zhu
- Department of Radiation Oncology, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medicine Sciences, Beijing, China.,National Center for Respiratory Disease, Beijing, China
| |
Collapse
|
23
|
Singh SS, Mattheolabakis G, Gu X, Withers S, Dahal A, Jois S. A grafted peptidomimetic for EGFR heterodimerization inhibition: Implications in NSCLC models. Eur J Med Chem 2021; 216:113312. [PMID: 33667849 PMCID: PMC8044046 DOI: 10.1016/j.ejmech.2021.113312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
Among the lung cancers, approximately 85% are histologically classified as non-small-cell lung cancer (NSCLC), a leading cause of cancer deaths worldwide. Epidermal growth factor receptors (EGFRs) are known to play a crucial role in lung cancer. HER2 overexpression is detected by immunohistochemistry in 2.4%-38% of NSCLC samples. EGFRs have been targeted with three generations of tyrosine kinase inhibitors (TKIs), and drug resistance has become a major issue; HER2 dimerization with EGFR also plays a major role in the development of resistance to TKI therapy. We have designed grafted peptides to bind to the HER2 extracellular domain (ECD) and inhibit protein-protein interactions of EGFR:HER2 and HER2:HER3. A sunflower trypsin inhibitor (SFTI-1) template was used to graft a peptidomimetic compound. Among several grafted peptides, SFTI-G5 exhibited antiproliferative activity in HER2-positive NSCLC cell lines such as Calu-3 cells with an IC50 value of 0.073 μM. SFTI-G5 was shown to bind to ECD of HER2 and inhibit EGFR:HER2 and HER2:HER3 dimerization and inhibit the phosphorylation of HER2 and downstream signaling proteins. As a proof-of-concept, the in vivo activity of SFTI-G5 was evaluated in two NSCLC mouse models. SFTI-G5 was able to inhibit tumor growth in both models. Furthermore, SFTI-G5 was shown to inhibit EGFR dimerization in tissue samples obtained from in vivo models. These grafted peptides can be used as novel dual inhibitors of EGFR dimerization in NSCLC.
Collapse
Affiliation(s)
- Sitanshu S Singh
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - George Mattheolabakis
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Xin Gu
- Department of Pathology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71103, USA
| | - Sita Withers
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Achyut Dahal
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Seetharama Jois
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA.
| |
Collapse
|
24
|
Türk S, Tok F, Erdoğan Ö, Çevik Ö, Tok TT, Koçyiğit-Kaymakçıoğlu B, Karakuş S. Synthesis, anticancer evaluation and in silico ADMET studies on urea/thiourea derivatives from gabapentin. PHOSPHORUS SULFUR 2021. [DOI: 10.1080/10426507.2020.1845678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Sevda Türk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkey
| | - Fatih Tok
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Ömer Erdoğan
- Department of Biochemistry, School of Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Özge Çevik
- Department of Biochemistry, School of Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Tuğba Taşkın Tok
- Department of Chemistry, Faculty of Sciences and Arts, Gaziantep University, Gaziantep, Turkey
| | | | - Sevgi Karakuş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| |
Collapse
|
25
|
Tancoš V, Farkašová A, Kviatkovská Z, Grendár M, Líšková A, Huťka Z, Plank L. Expression of programmed death-ligand 1 protein in pulmonary squamous cell carcinoma correlates with tumour necrosis but not with tumour differentiation. J Clin Pathol 2021; 75:373-378. [PMID: 33685938 DOI: 10.1136/jclinpath-2020-207171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/02/2021] [Accepted: 01/31/2021] [Indexed: 11/03/2022]
Abstract
AIMS Pulmonary squamous cell carcinoma (SqCC) represents the second most common non-small cell lung carcinoma type. The mechanisms which regulate programmed death ligand 1 (PD-L1) expression in this form of lung cancer are not fully elucidated yet. METHODS We immunohistochemically determined the level of PD-L1 expression using the Tumour Proportion Score system in surgical resections of 133 patients with pulmonary SqCC. The results from PD-L1 immunohistochemistry were analysed in relation to tumour differentiation and the presence of necrotic areas comprising at least 20% of the tumour mass. RESULTS No significant differences in terms of PD-L1 expression were found between SqCC subtypes as defined by the current WHO classification: better differentiated, keratinising tumours (12/24, 50.0 %) compared with less differentiated, non-keratinising and basaloid forms (62/109, 56.9 %) were PD-L1 positive in a comparable proportion of cases (p=0.1903). Contrary to that, SqCCs with the presence of necrosis (51/61, 83.6 %) had significantly more PD-L1-positive cases (p<0.001) compared with SqCCs without necrotic areas (23/72, 32.0 %) CONCLUSIONS: We demonstrated that PD-L1 expression in pulmonary SqCCs does not correlate with the traditionally defined degree of differentiation of these tumours. On the other hand, we found a significant association between the positive result of PD-L1 immunohistochemistry and tumour necrosis. Further investigation regarding the role of hypoxic pathways as presumable inducers of PD-L1 expression in pulmonary SqCCs might contribute to the understanding of this phenomenon.
Collapse
Affiliation(s)
- Vladimír Tancoš
- Univerzita Komenského v Bratislave Jesseniova Lekárska Fakulta v Martine, Martin, Slovakia
| | | | | | - Marián Grendár
- Univerzita Komenského v Bratislave Jesseniova Lekárska Fakulta v Martine, Martin, Slovakia
| | - Alena Líšková
- Univerzita Komenského v Bratislave Jesseniova Lekárska Fakulta v Martine, Martin, Slovakia
| | - Zdenko Huťka
- Univerzita Komenského v Bratislave Jesseniova Lekárska Fakulta v Martine, Martin, Slovakia
| | - Lukáš Plank
- Univerzita Komenského v Bratislave Jesseniova Lekárska Fakulta v Martine, Martin, Slovakia .,Martin's Biopsy Centre Ltd, Martin, Slovakia
| |
Collapse
|
26
|
Pulmonary administration of a CSF-1R inhibitor alters the balance of tumor-associated macrophages and supports first-line chemotherapy in a lung cancer model. Int J Pharm 2021; 598:120350. [PMID: 33545279 DOI: 10.1016/j.ijpharm.2021.120350] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/28/2021] [Accepted: 01/31/2021] [Indexed: 12/14/2022]
Abstract
Lung cancers remain the leading cause of cancer-related death in both men and women. Infiltrating immune cells in the tumor microenvironment (TME) play a critical role in the formation, progression, and the response of solid tumors to therapy, including in lung cancers. Clinical studies have established that tumor-associated macrophages (TAMs) and their phenotypical composition are critical immune infiltrates in the lung TME, with the abundance of the M2-like phenotype negatively correlating with patient survival. Colony-Stimulating Factor 1 (CSF-1) receptor (CSF-1R) is a type III protein tyrosine kinase receptor that plays an important role in the recruitment and differentiation of monocytes into tumor-promoting M2-like TAMs and their survival. In this work we evaluated the therapeutic potential of PLX 3397 (PLX), a small molecule CSF-1R inhibitor (CSF-1Ri), upon local lung administration in an immune-competent mouse model of lung cancer. The efficacy of local lung delivered PLX as single therapy was investigated first. As assessed by immunofluorescence of sections of lung tumor nodules, a statistically significant reduction in M2-like TAMs and an increase in M1-like TAMs was observed, thus leading to a shift in the (M1/M2) balance. Those changes in abundance of immune infiltrates correlated with a significant decrease in tumor burden when compared to control. When combined with systemically administered cisplatin (CIS) PLX treatment provided further benefits, leading to a significant decrease in tumor burden when compared to either PLX or CIS treatments alone, as measured by bioluminescence intensity (BLI) in vivo (thoracic area) and ex vivo (lung tissue). This combination therapy led to the most pronounced increase in M1/M2 ratio, followed by a significant decrease in M2-like TAMs with the CIS therapy. This work is clinically relevant as it demonstrates the potential of local lung administration of PLX to support standard of care chemotherapy for lung cancer management. This is important as the pulmonary route of administration is a plausible strategy for reducing the total dose of CSF-1Ris as the tissue of interest (lungs) can be locally targeted. Because the major off-target effect of CSF-1Ris is liver toxicity, reducing systemic concentration will support translation of those therapies, especially in combination with standard of care chemotherapy that has significant off-target toxicity and patient attrition itself. This work is scientifically relevant as we demonstrate for the first time that local administration of a CSF-1Ri to the lungs leads to a shift in the balance of TAMs in the TME of a model of lung tumor, adding to the sparse literature of CSF-1Ris related to lung cancers.
Collapse
|
27
|
Lee DS, Oh K. Cancer Stem Cells in the Immune Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1187:245-266. [PMID: 33983582 DOI: 10.1007/978-981-32-9620-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer stem cells are a subpopulation of cancer cells responsible for the most demanding and aggressive cancer cell phenotypes: therapy resistance, a self-protective feature of stem cells; distant metastasis, requiring anchorage independence for survival in the circulation; and recurrence, which is related to the dormant-active cycling of stem cells. Normal tissues are composed of parenchymal cells, supportive connective components, and cellular disposal systems for removing the products of physiological wear and tear. Cancer stem cells develop from normal counterparts and progressively interact with their microenvironments, modifying and conditioning the cancer microenvironment. Cancer-associated myeloid cells constitute a major element of the cancer microenvironment. During the process of carcinogenesis, cancer stem cells and their intimately associated myeloid cells mutually interact and evolve, such that the cancer cells potentiate the activity of the myeloid cells and, in return, the myeloid cells increase cancer stem cell characteristics. Normal myeloid cells function as key accessory cells to maintain homeostasis in normal tissues and organs; in cancers, these cells co-evolve with the malignant parenchymal cells and are involved in every aspect of cancer cell biology, including proliferation, invasion, distant metastasis, and the development of resistance to therapy. In this way, cancer-associated myeloid cells provide two of the key hallmarks of cancer: evasion of immune destruction and cancer-promoting inflammation.
Collapse
Affiliation(s)
- Dong-Sup Lee
- Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Keunhee Oh
- SillaJen, Inc., Seoul, Republic of Korea
| |
Collapse
|
28
|
Maharjan M, Tanvir RB, Chowdhury K, Duan W, Mondal AM. Computational identification of biomarker genes for lung cancer considering treatment and non-treatment studies. BMC Bioinformatics 2020; 21:218. [PMID: 33272232 PMCID: PMC7713218 DOI: 10.1186/s12859-020-3524-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Lung cancer is the number one cancer killer in the world with more than 142,670 deaths estimated in the United States alone in the year 2019. Consequently, there is an overreaching need to identify the key biomarkers for lung cancer. The aim of this study is to computationally identify biomarker genes for lung cancer that can aid in its diagnosis and treatment. The gene expression profiles of two different types of studies, namely non-treatment and treatment, are considered for discovering biomarker genes. In non-treatment studies healthy samples are control and cancer samples are cases. Whereas, in treatment studies, controls are cancer cell lines without treatment and cases are cancer cell lines with treatment. RESULTS The Differentially Expressed Genes (DEGs) for lung cancer were isolated from Gene Expression Omnibus (GEO) database using R software tool GEO2R. A total of 407 DEGs (254 upregulated and 153 downregulated) from non-treatment studies and 547 DEGs (133 upregulated and 414 downregulated) from treatment studies were isolated. Two Cytoscape apps, namely, CytoHubba and MCODE, were used for identifying biomarker genes from functional networks developed using DEG genes. This study discovered two distinct sets of biomarker genes - one from non-treatment studies and the other from treatment studies, each set containing 16 genes. Survival analysis results show that most non-treatment biomarker genes have prognostic capability by indicating low-expression groups have higher chance of survival compare to high-expression groups. Whereas, most treatment biomarkers have prognostic capability by indicating high-expression groups have higher chance of survival compare to low-expression groups. CONCLUSION A computational framework is developed to identify biomarker genes for lung cancer using gene expression profiles. Two different types of studies - non-treatment and treatment - are considered for experiment. Most of the biomarker genes from non-treatment studies are part of mitosis and play vital role in DNA repair and cell-cycle regulation. Whereas, most of the biomarker genes from treatment studies are associated to ubiquitination and cellular response to stress. This study discovered a list of biomarkers, which would help experimental scientists to design a lab experiment for further exploration of detail dynamics of lung cancer development.
Collapse
Affiliation(s)
- Mona Maharjan
- School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Raihanul Bari Tanvir
- School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Kamal Chowdhury
- School of Natural Sciences and Mathematics, Claflin University, Orangeburg, SC, USA
| | - Wenrui Duan
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Ananda Mohan Mondal
- School of Computing and Information Sciences, Florida International University, Miami, FL, USA.
| |
Collapse
|
29
|
Regulatory (FoxP3 +) T cells and TGF-β predict the response to anti-PD-1 immunotherapy in patients with non-small cell lung cancer. Sci Rep 2020; 10:18994. [PMID: 33149213 PMCID: PMC7642363 DOI: 10.1038/s41598-020-76130-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/01/2020] [Indexed: 01/28/2023] Open
Abstract
Antitumor immune responses induced by immune checkpoint inhibitors anti-PD-1 or anti-PD-L1 have been used as therapeutic strategies in advanced non-small cell lung cancer (NSCLC) patients over the last decade. Favorable antitumor activity to immune checkpoint inhibitors is correlated with high PD-L1 expression, increased tumor-infiltrating lymphocytes, and decreased suppressive immune cells including Treg cells, myeloid-derived suppressor cells, or tumor-associated macrophages in various cancer types. In this study, we investigated the potential correlation between clinical outcomes and peripheral blood immune cell profiles, specifically focused on FoxP3+ Treg cells, collected at baseline and one week after anti-PD-1 therapy in two independent cohorts of patients with NSCLC: a discovery cohort of 83 patients and a validation cohort of 49 patients. High frequencies of circulating Treg cells one week after anti-PD-1 therapy were correlated with a high response rate, longer progression-free survival, and overall survival. Furthermore, high levels of TGF-β and Treg cells were associated with favorable clinical outcomes. Our results suggest that higher levels of FoxP3+ Treg cells and TGF-β can predict a favorable response to anti-PD-1 immunotherapy in patients with advanced NSCLC.
Collapse
|
30
|
Tancoš V, Farkašová A, Kviatkovská Z, Grendár M, Líšková A, Huťka Z, Plank L. Non-small cell lung carcinomas with a minor sarcomatoid component and pleomorphic carcinomas are associated with high expression of programmed death ligand 1. Pathol Res Pract 2020; 216:153238. [PMID: 33059241 DOI: 10.1016/j.prp.2020.153238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 01/26/2023]
Abstract
Pleomorphic carcinomas are known to be highly programmed death ligand 1 (PD-L1) positive non-small cell lung cancer (NSCLC) types. However, the level of PD-L1 expression in lung carcinomas with a minor sarcomatoid component, comprising less than 10 % of the tumor mass, has not been determined yet. We hypothesized that NSCLC with a minor sarcomatoid component is more closely related to pleomorphic carcinomas in terms of PD-L1 expression than to NSCLC types without sarcomatoid features. The surgical resections from 690 lung carcinoma patients were retrospectively analyzed for the presence of PD-L1 by means of immunohistochemistry using the 22C3 PharmDx assay. The tumor proportion score system was applied to quantify the level of PD-L1 expression. Membranous staining present in ≥ 1 % of tumor cells was chosen as the cut-off to define a positive result for PD-L1 expression. Tumors were allocated into one of four subgroups: "adenocarcinoma", "squamous cell carcinoma", "pleomorphic carcinoma", or "NSCLC with a minor sarcomatoid component". PD-L1 expression in pleomorphic carcinomas (26/32, 81.3 %) and in the subgroup of NSCLC with a minor sarcomatoid component (35/46, 76.1 %) was identified in a comparable proportion of cases. Pleomorphic carcinomas were significantly more often PD-L1 positive than adenocarcinomas (p < 0.001) or squamous cell carcinomas (p = 0.0015). Accordingly, the proportion of PD-L1 expressing NSCLC with a minor sarcomatoid component was significantly higher than that of the adenocarcinoma (p < 0.001) or squamous cell carcinoma (p = 0.002) subgroup. In summary, we identified a presumable new subgroup of highly PD-L1 positive neoplasms within the NSCLC spectrum that is related to pleomorphic carcinomas in terms of PD-L1 expression. Further investigation regarding genetic relation and mechanism of PD-L1 expression in these two NSCLC categories is recommended.
Collapse
Affiliation(s)
- Vladimír Tancoš
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Kollárova 2, 03659, Martin, Slovakia.
| | - Anna Farkašová
- Martin's Biopsy Centre Ltd., Prieloztek 1, 03601, Martin, Slovakia.
| | | | - Marián Grendár
- Department of Bioinformatics, Biomedical Centre Martin, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Mala Hora 4C, 03601, Martin, Slovakia.
| | - Alena Líšková
- Clinic of Obstetrics and Gynecology, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Kollárova 2, 03601, Martin, Slovakia.
| | - Zdenko Huťka
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Kollárova 2, 03659, Martin, Slovakia.
| | - Lukáš Plank
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Kollárova 2, 03659, Martin, Slovakia; Martin's Biopsy Centre Ltd., Prieloztek 1, 03601, Martin, Slovakia.
| |
Collapse
|
31
|
Geng Q, Rohondia SO, Khan HJ, Jiao P, Dou QP. Small molecules as antagonists of co-inhibitory pathways for cancer immunotherapy: a patent review (2018-2019). Expert Opin Ther Pat 2020; 30:677-694. [PMID: 32715813 DOI: 10.1080/13543776.2020.1801640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Therapeutic antibodies blocking co-inhibitory pathways do not attack tumor cells directly, but instead bind to their targeted proteins and mobilize the immune system to eradicate tumors. However, only a small fraction of patients with certain cancer types can benefit from the antibodies. Additionally, antibodies have shown serious immune-related adverse events in certain patients. Small-molecule antagonists may be a complementary and potentially synergistic approach to antibodies for patients with various cancers. AREAS COVERED The authors review the small molecules as antagonists of co-inhibitory pathway proteins, summarize their preliminary SARs, discuss biochemistry assays used in patents for the development of small molecules as novel antagonists. EXPERT OPINION The disclosed pharmacophores of small molecules as co-inhibitory pathway antagonists are represented by biphenyl derivatives, biaryl derivatives, teraryl derivatives, quateraryl derivatives, and oxadiazole/thiadiazole derivatives. However, these antagonists are still inferior to therapeutic antibodies in their inhibitory activities due to relatively flat of human co-inhibitory pathways proteins. Allosteric modulators may be an alternative approach. The more safety and efficacy evaluation trials of small-molecule antagonists targeting co-inhibitory pathways should be performed to demonstrate the proof-of-principle that small-molecule antagonists can result in sustained safety and antitumor response in the near future.
Collapse
Affiliation(s)
- Qiaohong Geng
- Department of Chemistry, Qilu Normal University , Jinan, China
| | - Sagar O Rohondia
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University , Detroit, MI, USA
| | - Harras J Khan
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University , Detroit, MI, USA
| | - Peifu Jiao
- Department of Chemistry, Qilu Normal University , Jinan, China
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University , Detroit, MI, USA
| |
Collapse
|
32
|
Pu JT, Hu Z, Zhang DG, Zhang T, He KM, Dai TY. MiR-654-3p Suppresses Non-Small Cell Lung Cancer Tumourigenesis by Inhibiting PLK4. Onco Targets Ther 2020; 13:7997-8008. [PMID: 32884289 PMCID: PMC7431606 DOI: 10.2147/ott.s258616] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/25/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose MiR-654-3p plays important roles in many types of malignant tumours. However, the biological function of miR-654-3p in non-small cell lung cancer (NSCLC) remains unknown. In this study, the role of miR-654-3p in NSCLC was investigated. Methods qRT-PCR was used to evaluate the level of miR-654-3p in NSCLC tissues and cell lines, while Cell Counting Kit-8, Annexin V/propidium iodide dual staining or TUNEL staining were used to investigate proliferation and apoptosis of NSCLC cells. Luciferase assays and Western blotting were performed to validate potential targets of miR-654-3p. Results MiR-654-3p levels were significantly decreased in NSCLC patients and cell lines and were significantly correlated with the tumour size and tumour node metastasis stage of NSCLC patients. In A549 cells, miR-654-3p overexpression significantly increased apoptosis and inhibited growth both in vivo and in vitro, while downregulation of miR-654-3p had the opposite effects. In addition, polo-like kinase 4 (PLK4) was shown to be a target gene of miR-654-3p that is negatively regulated by miR-654-3p in A549 cells. Furthermore, PLK4 was observed to be highly expressed in NSCLC tissues and cells, and PLK4 overexpression abolished the inhibitory effects of miR-654-3p overexpression on NSCLC cell proliferation. Finally, the animal experiment results further demonstrated that miR-654-3p inhibits tumour growth and regulates PLK4 expression. Conclusion Our results demonstrate that miR-654-3p functions as a growth-suppressing miRNA by targeting PLK4 in NSCLC.
Collapse
Affiliation(s)
- Jiang-Tao Pu
- Department of Thoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Zhi Hu
- Department of Thoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Deng-Guo Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Tao Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Kai-Ming He
- Department of Thoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Tian-Yang Dai
- Department of Thoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
33
|
Ramelow J, Brooks CD, Gao L, Almiman AA, Williams TM, Villalona-Calero MA, Duan W. The oncogenic potential of a mutant TP53 gene explored in two spontaneous lung cancer mice models. BMC Cancer 2020; 20:738. [PMID: 32770960 PMCID: PMC7414707 DOI: 10.1186/s12885-020-07212-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/23/2020] [Indexed: 12/25/2022] Open
Abstract
Background Lung cancer is the number one cancer killer worldwide. A major drawback in the lung cancer treatment field is the lack of realistic mouse models that replicate the complexity of human malignancy and immune contexture within the tumor microenvironment. Such models are urgently needed. Mutations of the tumor protein p53 are among the most common alterations in human lung cancers. Methods Previously, we developed a line of lung cancer mouse model where mutant human TP53-273H is expressed in a lung specific manner in FVB/N background. To investigate whether the human TP53 mutant has a similar oncogenic potential when it is expressed in another strain of mouse, we crossed the FVB/N-SPC-TP53-273H mice to A/J strain and created A/J-SPC-TP53-273H transgenic mice. We then compared lung tumor formation between A/J-SPC-TP53-273H and FVB/N-SPC-TP53-273H. Results We found the TP53-273H mutant gene has a similar oncogenic potential in lung tumor formation in both mice strains, although A/J strain mice have been found to be a highly susceptible strain in terms of carcinogen-induced lung cancer. Both transgenic lines survived more than 18 months and developed age related lung adenocarcinomas. With micro CT imaging, we found the FVB-SPC-TP53-273H mice survived more than 8 weeks after initial detection of lung cancer, providing a sufficient window for evaluating new anti-cancer agents. Conclusions Oncogenic potential of the most common genetic mutation, TP53-273H, in human lung cancer is unique when it is expressed in different strains of mice. Our mouse models are useful tools for testing novel immune checkpoint inhibitors or other therapeutic strategies in the treatment of lung cancer.
Collapse
Affiliation(s)
- Julian Ramelow
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA.,Biomolecular Sciences Institute, The Florida International University, Miami, Florida, 33199, USA.,Biological Sciences, College of Arts, Science and Education, The Florida International University, Miami, Florida, 33199, USA
| | - Christopher D Brooks
- Comprehensive Cancer Center at the Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Li Gao
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA
| | - Abeer A Almiman
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA
| | - Terence M Williams
- Comprehensive Cancer Center at the Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | | | - Wenrui Duan
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA. .,Biomolecular Sciences Institute, The Florida International University, Miami, Florida, 33199, USA. .,Comprehensive Cancer Center at the Ohio State University College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
34
|
Koh J, Kim Y, Lee KY, Hur JY, Kim MS, Kim B, Cho HJ, Lee YC, Bae YH, Ku BM, Sun JM, Lee SH, Ahn JS, Park K, Ahn MJ. MDSC subtypes and CD39 expression on CD8 + T cells predict the efficacy of anti-PD-1 immunotherapy in patients with advanced NSCLC. Eur J Immunol 2020; 50:1810-1819. [PMID: 32510574 PMCID: PMC7689686 DOI: 10.1002/eji.202048534] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/02/2020] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
The major suppressive immune cells in tumor sites are myeloid derived suppressor cells (MDSCs), tumor‐associated macrophages (TAMs), and Treg cells, and the major roles of these suppressive immune cells include hindering T‐cell activities and supporting tumor progression and survival. In this study, we analyzed the pattern of circulating MDSC subtypes in patients with non‐small cell lung cancer (NSCLC) whether those suppressive immune cells hinder T‐cell activities leading to poor clinical outcomes. First, we verified PMN‐MDSCs, monocytic‐MDSCs (M‐MDSCs), and Treg cells increased according to the stages of NSCLC, and MDSCs effectively suppressed T‐cell activities and induced T‐cell exhaustion. The analysis of NSCLC patients treated with anti‐PD‐1 immunotherapy demonstrated that low PMN‐MDSCs, M‐MDSCs, and CD39+CD8+ T cells as an individual and all together were associated with longer progression free survival and overall survival, suggesting PMN‐MDSCs, M‐MDSCs, and CD39+CD8+ T cells frequencies in peripheral blood might be useful as potential predictive and prognostic biomarkers.
Collapse
Affiliation(s)
- Jiae Koh
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Youjin Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Division of Hematology-Oncology, Department of Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Kyoung Young Lee
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Young Hur
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mi Soon Kim
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Boram Kim
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Jin Cho
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeong Chan Lee
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Yeon Hee Bae
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Bo Mi Ku
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se-Hoon Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Ju Ahn
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Tancoš V, Grendár M, Farkašová A, Huťka Z, Mičák J, Kviatkovská Z, Hardman TC, Hardy GAD, Plank L. Programmed death ligand 1 protein expression, histological tumour differentiation and intratumoural heterogeneity in pulmonary adenocarcinoma. Pathology 2020; 52:538-545. [PMID: 32586689 DOI: 10.1016/j.pathol.2020.03.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 01/15/2023]
Abstract
Intratumoural heterogeneity of pulmonary adenocarcinoma challenges the accurate interpretation of programmed death ligand 1 (PD-L1) immunohistochemistry, which is the only validated predictive marker for successful anti-PD-1/PD-L1 immunotherapy. The aim of this study was to determine whether PD-L1 expression is related to adenocarcinoma histological differentiation in a retrospective analysis of tumour biopsies with intratumoural histological heterogeneity. Adenocarcinomas with high intratumoural heterogeneity were categorised as 'mixed adenocarcinomas'. PD-L1 expression was determined immunohistochemically using tumour proportion scores (TPS). In 'mixed adenocarcinomas' PD-L1 scores were assessed across tumour areas with specific histological patterns. Comparisons were performed between histologically distinct differentiated tumours and/or histological areas. Poorly differentiated adenocarcinomas, represented by predominantly solid or micropapillary histological patterns, showed significantly higher expression of PD-L1 than other subtypes (p<0.001). Differentiation of intra-adenocarcinoma components was inversely correlated with PD-L1 expression: there were more PD-L1 positive cells in poorly differentiated areas than less differentiated (p<0.001), or than well differentiated areas (p<0.001), and in less differentiated more than well differentiated areas (p=0.001). In conclusion, PD-L1 expression is associated with poorly differentiated morphology in adenocarcinomas with intratumoural histological heterogeneity. Consequently, a TPS approach may not account for the contribution of more aggressive tumour components with higher levels of PD-L1 expression in within the tumour. Performing spectral analyses of PD-L1 expression across tumours is likely to be more accurate.
Collapse
Affiliation(s)
- Vladimír Tancoš
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Slovakia.
| | - Marián Grendár
- Department of Bioinformatics, Biomedical Centre Martin, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia
| | | | - Zdenko Huťka
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Slovakia
| | - Jozef Mičák
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Slovakia
| | | | | | | | - Lukáš Plank
- Department of Pathological Anatomy, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital in Martin, Slovakia; Martin's Biopsy Centre Ltd, Martin, Slovakia
| |
Collapse
|
36
|
Li N, Zheng H, Zheng B, Chen C, Cai H, Liu M. Economic Evaluations of Immune Checkpoint Inhibitors for Patients with Non-Small Cell Lung Cancer: A Systematic Review. Cancer Manag Res 2020; 12:4503-4518. [PMID: 32606944 PMCID: PMC7297344 DOI: 10.2147/cmar.s248020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/09/2020] [Indexed: 12/26/2022] Open
Abstract
Objective This review aimed to assess the quality of available evidence on the economic evaluations of immune checkpoint inhibitors in patients with non-small cell lung cancer (NSCLC) and provide evidence to improve the efficiency of healthcare resources. Materials and Methods Literature search was performed using some electronic databases (PubMed, Embase and Cochrane Central Register of Controlled Trials). Final search was performed in December 2019. Study characteristics and results were recorded and compared. The quality of the studies was assessed using the Consolidated Health Economic Evaluation Reporting Standards (CHEERS) checklists. We did not elaborate the restrictions on the target population. We included patients with squamous or non-squamous NSCLC and metastatic or advanced cancer. Results Of 98 papers considered, 21 were chosen for this review. Most of them are cost-effectiveness analysis. Comparative regimens consisted of either immune checkpoint inhibitor monotherapy, immune checkpoint inhibitor plus chemotherapy, or chemotherapy alone. Fourteen, four, and three studies were about pembrolizumab, nivolumab, and atezolizumab, respectively. The methods mostly used in these studies were modeling and sensitivity analysis. All studies used quality-adjusted life year (QALY) and life years (LY) as outcomes. Most studies were conducted in high-income countries. Based on the willingness to pay threshold, atezolizumab, and pembrolizumab were found to be cost-effective in one and 10 studies, respectively. None of the studies concluded that nivolumab was cost-effective. For quality assessment, all studies fulfilled more than 50% of the CHEERS checklist. Conclusion The included studies indicated that pembrolizumab regimens are cost-effective as first-line treatment for patients with NSCLC in developed countries. Nivolumab and atezolizumab are likely to be cost-effective as second-line treatment but not as first-line treatment.
Collapse
Affiliation(s)
- Na Li
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China.,School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Huanrui Zheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China.,School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Bin Zheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China.,School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Chaoxin Chen
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China.,School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Hongfu Cai
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China.,School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China.,School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| |
Collapse
|
37
|
Youn JI, Park SM, Park S, Kim G, Lee HJ, Son J, Hong MH, Ghaderpour A, Baik B, Islam J, Choi JW, Lee EY, Kim HR, Seo SU, Paik S, Yoon HI, Jung I, Xin CF, Jin HT, Cho BC, Seong SY, Ha SJ, Kim HR. Peripheral natural killer cells and myeloid-derived suppressor cells correlate with anti-PD-1 responses in non-small cell lung cancer. Sci Rep 2020; 10:9050. [PMID: 32493990 PMCID: PMC7270107 DOI: 10.1038/s41598-020-65666-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/23/2020] [Indexed: 12/21/2022] Open
Abstract
Inhibition of immune checkpoint proteins like programmed death 1 (PD-1) is a promising therapeutic approach for several cancers, including non-small cell lung cancer (NSCLC). Although PD-1 ligand (PD-L1) expression is used to predict anti-PD-1 therapy responses in NSCLC, its accuracy is relatively less. Therefore, we sought to identify a more accurate predictive blood biomarker for evaluating anti-PD-1 response. We evaluated the frequencies of T cells, B cells, natural killer (NK) cells, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), mononuclear myeloid-derived suppressor cells (M-MDSCs), and Lox-1+ PMN-MDSCs in peripheral blood samples of 62 NSCLC patients before and after nivolumab treatment. Correlation of immune-cell population frequencies with treatment response, progression-free survival, and overall survival was also determined. After the first treatment, the median NK cell percentage was significantly higher in responders than in non-responders, while the median Lox-1+ PMN-MDSC percentage showed the opposite trend. NK cell frequencies significantly increased in responders but not in non-responders. NK cell frequency inversely correlated with that of Lox-1+ PMN-MDSCs after the first treatment cycle. The NK cell-to-Lox-1+ PMN-MDSC ratio (NMR) was significantly higher in responders than in non-responders. Patients with NMRs ≥ 5.75 after the first cycle had significantly higher objective response rates and longer progression-free and overall survival than those with NMRs <5.75. NMR shows promise as an early predictor of response to further anti-PD-1 therapy.
Collapse
Affiliation(s)
- Je-In Youn
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea.
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea.
- Research Institute, ProGen, Inc., Seongnam-si, Gyeonggi-do, Korea.
| | - Su-Myeong Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Seyeon Park
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Gamin Kim
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Hee-Jae Lee
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea
| | - Jimin Son
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Min Hee Hong
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Aziz Ghaderpour
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Bumseo Baik
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jahirul Islam
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Woong Choi
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea
| | - Eun-Young Lee
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea
| | - Hang-Rae Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Uk Seo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea
| | - Soonmyung Paik
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hong In Yoon
- Yonsei Cancer Center, Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Inkyung Jung
- Department of Biostatistics and Medical Informatics, Yonsei University College of Medicine, Seoul, Korea
| | - Chun-Feng Xin
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi-City, Kyungbuk, Korea
| | - Hyun-Tak Jin
- Research Institute, ProGen, Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Byoung Chul Cho
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi-City, Kyungbuk, Korea
| | - Seung-Yong Seong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea.
| | - Hye Ryun Kim
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
38
|
Kim KH, Kim CG, Shin EC. Peripheral blood immune cell-based biomarkers in anti-PD-1/PD-L1 therapy. Immune Netw 2020; 20:e8. [PMID: 32158596 PMCID: PMC7049582 DOI: 10.4110/in.2020.20.e8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint blockade targeting PD-1 and PD-L1 has resulted in unprecedented clinical benefit for cancer patients. Anti-PD-1/PD-L1 therapy has become the standard treatment for diverse cancer types as monotherapy or in combination with other anti-cancer therapies, and its indications are expanding. However, many patients do not benefit from anti-PD-1/PD-L1 therapy due to primary and/or acquired resistance, which is a major obstacle to broadening the clinical applicability of anti-PD-1/PD-L1 therapy. In addition, hyperprogressive disease, an acceleration of tumor growth following anti-PD-1/PD-L1 therapy, has been proposed as a new response pattern associated with deleterious prognosis. Anti-PD-1/PD-L1 therapy can also cause a unique pattern of adverse events termed immune-related adverse events, sometimes leading to treatment discontinuation and fatal outcomes. Investigations have been carried out to predict and monitor treatment outcomes using peripheral blood as an alternative to tissue biopsy. This review summarizes recent studies utilizing peripheral blood immune cells to predict various outcomes in cancer patients treated with anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Chang Gon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
39
|
Oh SJ, Lee J, Kim Y, Song KH, Cho E, Kim M, Jung H, Kim TW. Far Beyond Cancer Immunotherapy: Reversion of Multi-Malignant Phenotypes of Immunotherapeutic-Resistant Cancer by Targeting the NANOG Signaling Axis. Immune Netw 2020; 20:e7. [PMID: 32158595 PMCID: PMC7049583 DOI: 10.4110/in.2020.20.e7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer immunotherapy, in the form of vaccination, adoptive cellular transfer, or immune checkpoint inhibitors, has emerged as a promising practice within the field of oncology. However, despite the developing field's potential to revolutionize cancer treatment, the presence of immunotherapeutic-resistant tumor cells in many patients present a challenge and limitation to these immunotherapies. These cells not only indicate immunotherapeutic resistance, but also show multi-modal resistance to conventional therapies, abnormal metabolism, stemness, and metastasis. How can immunotherapeutic-resistant tumor cells render multi-malignant phenotypes? We reasoned that the immune-refractory phenotype could be associated with multi-malignant phenotypes and that these phenotypes are linked together by a factor that acts as the master regulator. In this review, we discussed the role of the embryonic transcription factor NANOG as a crucial master regulator we named “common factor” in multi-malignant phenotypes and presented strategies to overcome multi-malignancy in immunotherapeutic-resistant cancer by restraining the NANOG-mediated multi-malignant signaling axis. Strategies that blunt the NANOG axis could improve the clinical management of therapy-refractory cancer.
Collapse
Affiliation(s)
- Se Jin Oh
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Jaeyoon Lee
- College of Science, College of Social Sciences and Humanities, Northeastern University, Boston, MA 02115, USA
| | - Yukang Kim
- Korea University College of Medicine, Seoul 02841, Korea
| | - Kwon-Ho Song
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Eunho Cho
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Minsung Kim
- Korea University College of Medicine, Seoul 02841, Korea
| | - Heejae Jung
- Korea University College of Medicine, Seoul 02841, Korea
| | - Tae Woo Kim
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
40
|
Niu C, Li M, Zhu S, Chen Y, Zhou L, Xu D, Xu J, Li Z, Li W, Cui J. PD-1-positive Natural Killer Cells have a weaker antitumor function than that of PD-1-negative Natural Killer Cells in Lung Cancer. Int J Med Sci 2020; 17:1964-1973. [PMID: 32788875 PMCID: PMC7415385 DOI: 10.7150/ijms.47701] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
Antibodies targeting the immune checkpoint inhibitor, programmed cell death 1 (PD-1), have provided a breakthrough in the treatment of lung cancer. However, the function of PD-1 in natural killer (NK) cells of cancer patients remains unclear. Herein, we analyzed the expression of PD-1 on the NK cells in the peripheral blood of patients with lung cancer and found that the level of PD-1+ NK cells in patients was significantly higher than that in healthy individuals. Moreover, these PD-1+ NK cells demonstrated a weaker ability to secrete interferon-gamma (INF-γ), granzyme B, and perforin, and exhibited lower CD107a expression. Importantly, in patients with lung cancer, the percentage of PD-1+ NK cells was significantly positively correlated with the concentration of IL-2 in the plasma, which was also higher than that in healthy individuals. In addition, IL-2 could increase the expression of PD-1 on NK cells in vitro, indicating that high IL-2 level in the plasma is largely responsible for the abundance of PD-1+ NK cells in patients with lung cancer. These findings demonstrate intriguing mechanisms for understanding the expression of PD-1 on NK cells and the function of PD-1+ NK cells in lung cancer. This study confirms and extends previous studies demonstrating that PD-1 can negatively regulate the antitumor function of NK cells.
Collapse
Affiliation(s)
- Chao Niu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Min Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan Zhu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Yongchong Chen
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Lei Zhou
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Dongsheng Xu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Jianting Xu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhaozhi Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Wei Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Jiuwei Cui
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
41
|
Hauber B, Penrod JR, Gebben D, Musallam L. The Value of Hope: Patients' and Physicians' Preferences for Survival in Advanced Non-Small Cell Lung Cancer. Patient Prefer Adherence 2020; 14:2093-2104. [PMID: 33154633 PMCID: PMC7608144 DOI: 10.2147/ppa.s248295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 09/19/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Immuno-oncology treatments offer patients with advanced non-small cell lung cancer (NSCLC) treatment options with greater probability of durable survival and a different toxicity profile compared with traditional chemotherapy. The objective of this study was to explore the importance of increases in the probability of long-term survival versus changes in expected (median) survival and treatment toxicities among patients with advanced NSCLC and physicians. PATIENTS AND METHODS In a discrete-choice experiment, oncologists and patients diagnosed with NSCLC chose between profiles of treatments for advanced NSCLC offering different combinations of benefits (expected, best-case, and worst-case survival) and risks. We analyzed preference data from each sample using a random-parameters logit model that controls for preference heterogeneity and the panel nature of the data. RESULTS Both patients and physicians expressed a strong preference for improving the probability of best-case survival; however, patients viewed increases in the probability of long-term survival as more important than increases in expected survival, while the opposite was true for physicians. Both patients and physicians weighted survival to be more important than toxicities. CONCLUSION This study identified a potentially important divergence between physician and patient perspectives on survival statistics. Physicians placed more importance on increases in expected survival than did patients with NSCLC. The importance patients placed on long-term survival reinforce previous research identifying the primacy of hope as a value among seriously ill patients. The findings underscore the importance of considering patients' priorities and in shared decision-making when choosing treatment.
Collapse
Affiliation(s)
- Brett Hauber
- RTI Health Solutions, Research Triangle Park, NC, USA
- Correspondence: Brett Hauber Email
| | | | - David Gebben
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | |
Collapse
|
42
|
Huang M, Lopes GDL, Insinga RP, Burke T, Ejzykowicz F, Zhang Y, Feliciano JL. Cost–effectiveness of pembrolizumab versus chemotherapy as first-line treatment in PD-L1-positive advanced non-small-cell lung cancer in the USA. Immunotherapy 2019; 11:1463-1478. [DOI: 10.2217/imt-2019-0178] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: This analysis aimed to evaluate the cost–effectiveness of pembrolizumab monotherapy as first-line treatment in advanced non-small-cell lung cancer patients with a programmed death ligand 1 (PD-L1) tumor proportion score ≥1% from a US payer perspective. Materials & methods: A partitioned survival model was developed using efficacy and safety data from the KEYNOTE-042 trial and projected over 20 years. Costs accounted for treatment, toxicity and disease management. Quality-adjusted life-years (QALYs) and incremental cost–effectiveness ratios were reported. Results: Pembrolizumab resulted in an expected gain of 0.60 life years and 0.49 QALYs compared with platinum-based chemotherapy. The incremental cost–effectiveness ratio was US$130,155/QALY. Conclusion: Pembrolizumab is projected to be cost-effective compared with platinum-based chemotherapy as first-line treatment for advanced non-small-cell lung cancer with PD-L1 tumor proportion score ≥1%.
Collapse
Affiliation(s)
- Min Huang
- Center for Observational & Real World Evidence (CORE), Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Gilberto de Lima Lopes
- Sylvester Comprehensive Cancer Center, University of Miami & the Miller School of Medicine, Miami, FL 33136, USA
| | - Ralph P Insinga
- Center for Observational & Real World Evidence (CORE), Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Thomas Burke
- Center for Observational & Real World Evidence (CORE), Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Flavia Ejzykowicz
- Center for Observational & Real World Evidence (CORE), Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Ying Zhang
- HTA Statistics Europe, Merck Sharp & Dohme, 1200 Brussels, Belgium
| | - Josephine L Feliciano
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
43
|
Zhang Q, Zhang J, Zhang J, Aerxiding P, Quhai A, Chen C, Shan L. The Biological-Behavioral Effect Of Neuritin On Non-Small Cell Lung Cancer Vascular Endothelial Cells Via VEGFR And Notch1. Onco Targets Ther 2019; 12:9747-9755. [PMID: 31819478 PMCID: PMC6876221 DOI: 10.2147/ott.s212771] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/06/2019] [Indexed: 12/30/2022] Open
Abstract
Purpose This study aims to elucidate the biological behavior of Neuritin abnormal expression in pulmonary vascular endothelial cells (VECs) of non-small cell lung cancer (NSCLC), and explore its possible underlying mechanisms. Patients and methods Primary NSCLC-VECs were isolated from 10 cancer tissues from NSCLC patients, purified and identified by CD34 and Factor VIII staining. Real-time PCR and Western-blot were adopted for detecting the expression levels of Neuritin, Notch1, and VEGFR in NSCLC-VECs and HPMECs. Neuritin-overexpression, Neuritin-knockdown NSCLC-VECs and HPMECs were constructed by transfection of pcDNA3, 1-Neuritin vector, and pBS/U6-Neuritin siRNA. Changes in cell proliferation, migration, cell cycle, and apoptosis were determined by using the MTT assay, scratch assay, transwell migration assay, and flow cytometry, respectively. Post-transfection changes in cell morphology were examined by scanning electron microscopy. Results The expression of Neuritin in NSCLC-VECs was significantly higher compared to that in HPMECs (p<0.01). Overexpression of Neuritin increased the expression of VEGFR while it reduced the expression of Notch1 (p<0.01); it also promoted cell proliferation, scratch healing, and in vitro migration (p<0.05) in HPMECs and NSCLC-VECs cells. Additionally, overexpression of Neuritin stimulated cell cycle progression and inhibited apoptosis in HPMECs and NSCLC-VECs (p<0.001). Under electron microscope, the pseudopodium of cell surface was obvious, indicating that the intercellular adhesion was upregulated. However, knockdown of Neuritin in HPMECs and NSCLC-VECs played exactly the opposite roles. Conclusion Neuritin was key in the progression of NSCLC through its biological activities, including anti-apoptosis, promoting VEC proliferation, migration, and cell cycle progression. Neuritin may affect its biological activity by positively regulating VEGFR expression and negatively regulating Notch1 signaling. Neuritin may serve as a potential biomarker for NSCLC.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Thoraciconcology, The Third Affiliated Hospital of Xinjiang Medical University, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, People's Republic of China
| | - Juan Zhang
- Department of Respiratory and Critical Care Medicine, First People's Hospital of Kashgar, Xinjiang 844000, People's Republic of China
| | - Jian Zhang
- Health Corps of the People's Liberation Army 69260 Troops, Urumqi, Xinjiang, 830002, People's Republic of China
| | - Patiguli Aerxiding
- Department of Thoraciconcology, The Third Affiliated Hospital of Xinjiang Medical University, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, People's Republic of China
| | - Amina Quhai
- Department of Thoraciconcology, The Third Affiliated Hospital of Xinjiang Medical University, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, People's Republic of China
| | - Cuncun Chen
- Department of Thoracic Surgery, Chest Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830049, People's Republic of China
| | - Li Shan
- Department of Thoraciconcology, The Third Affiliated Hospital of Xinjiang Medical University, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, People's Republic of China
| |
Collapse
|
44
|
Li Y, Ding K, Hu X, Wu L, Zhou D, Rao M, Lin N, Zhang C. DYRK1A inhibition suppresses STAT3/EGFR/Met signalling and sensitizes EGFR wild-type NSCLC cells to AZD9291. J Cell Mol Med 2019; 23:7427-7437. [PMID: 31454149 PMCID: PMC6815810 DOI: 10.1111/jcmm.14609] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/14/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
DYRK1A is considered a potential cancer therapeutic target, but the role of DYRK1A in NSCLC oncogenesis and treatment requires further investigation. In our study, high DYRK1A expression was observed in tumour samples from patients with lung cancer compared with normal lung tissues, and the high levels of DYRK1A were related to a reduced survival time in patients with lung cancer. Meanwhile, the DYRK1A inhibitor harmine could suppress the proliferation of NSCLC cells compared to that of the control. As DYRK1A suppression might be effective in treating NSCLC, we next explored the possible specific molecular mechanisms that were involved. We showed that DYRK1A suppression by siRNA could suppress the levels of EGFR and Met in NSCLC cells. Furthermore, DYRK1A siRNA could inhibit the expression and nuclear translocation of STAT3. Meanwhile, harmine could also regulate the STAT3/EGFR/Met signalling pathway in human NSCLC cells. AZD9291 is effective to treat NSCLC patients with EGFR-sensitivity mutation and T790 M resistance mutation, but the clinical efficacy in patients with wild-type EGFR remains modest. We showed that DYRK1A repression could enhance the anti-cancer effect of AZD9291 by inducing apoptosis and suppressing cell proliferation in EGFR wild-type NSCLC cells. In addition, harmine could enhance the anti-NSCLC activity of AZD9291 by modulating STAT3 pathway. Finally, harmine could enhance the anti-cancer activity of AZD9291 in primary NSCLC cells. Collectively, targeting DYRK1A might be an attractive target for AZD9291 sensitization in EGFR wild-type NSCLC patients.
Collapse
Affiliation(s)
- Yang‐ling Li
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou, ZhejiangChina
| | - Ke Ding
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou, ZhejiangChina
| | - Xiu Hu
- School of MedicineZhejiang University City CollegeHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou, ZhejiangChina
| | - Lin‐wen Wu
- School of MedicineZhejiang University City CollegeHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou, ZhejiangChina
| | - Dong‐mei Zhou
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou, ZhejiangChina
| | - Ming‐jun Rao
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou, ZhejiangChina
| | - Neng‐ming Lin
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou, ZhejiangChina
- Hangzhou Translational Medicine Research Center, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou, ZhejiangChina
| | - Chong Zhang
- School of MedicineZhejiang University City CollegeHangzhouZhejiangChina
| |
Collapse
|
45
|
Bridges JF, la Cruz MD, Pavilack M, Flood E, Janssen EM, Chehab N, Fernandes AW. Patient preferences for attributes of tyrosine kinase inhibitor treatments for EGFR mutation-positive non-small-cell lung cancer. Future Oncol 2019; 15:3895-3907. [PMID: 31621403 DOI: 10.2217/fon-2019-0396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: EGFR-tyrosine kinase inhibitors (TKIs) vary in efficacy, side effects (SEs) and dosing regimen. We explored EGFR-TKI treatment attribute preferences in EGFR mutation-positive metastatic non-small-cell lung cancer. Materials & methods: Patients completed a survey utilizing preference elicitation methods: direct elicitation of four EGFR-TKI profiles describing progression-free survival (PFS), severe SE risk, administration; discrete choice experiment involving 12 choice tasks. Results: 90 participated. The preferred profile (selected 89% of times) had the longest PFS (18 months) and the lowest severe SE risk (5%). Patients would need compensation with ≥three-times longer PFS for severe SEs. Patients would accept ≤7 months PFS reduction for oral treatments versus intravenous. Conclusion: Patients preferred longer PFS but were willing to accept reduced PFS for more favorable SEs and dosing convenience.
Collapse
Affiliation(s)
- John Fp Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | | | | | - Emuella Flood
- Patient Centered Outcomes, ICON, Gaithersburg, MD 20807, USA.,AstraZeneca, Gaithersburg, MD 20807, USA
| | - Ellen M Janssen
- Patient Centered Outcomes, ICON, Gaithersburg, MD 20807, USA
| | | | | |
Collapse
|
46
|
Yu S, Yang D, Ye Y, Liu P, Chen Z, Lei T, Pu J, Liu L, Wang Z. Long noncoding RNA actin filament-associated protein 1 antisense RNA 1 promotes malignant phenotype through binding with lysine-specific demethylase 1 and repressing HMG box-containing protein 1 in non-small-cell lung cancer. Cancer Sci 2019; 110:2211-2225. [PMID: 31069893 PMCID: PMC6609801 DOI: 10.1111/cas.14039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/28/2019] [Accepted: 05/06/2019] [Indexed: 12/16/2022] Open
Abstract
The number of documented long noncoding RNAs (lncRNAs) has dramatically increased, and their biological functions and underlying mechanisms in pathological processes, especially cancer, remain to be elucidated. Actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) is a 6810-nt lncRNA located on chromosome 4p16.1 that was first reported to be upregulated in esophageal adenocarcinoma tissues and cell lines. Here we reported that AFAP1-AS1, recruiting and binding to lysine-specific demethylase 1 (LSD1), was generally overexpressed in human non-small-cell lung cancer (NSCLC) tissues using quantitative real-time PCR. Higher AFAP1-AS1 expression was significantly correlated with larger tumor size (P = .008), lymph node metastasis (P = .025), higher TNM stage (P = .024), and worse overall survival in NSCLC patients. In vitro experiments revealed that AFAP1-AS1 downregulation inhibited cell migration and induced apoptosis; AFAP1-AS1 knockdown also hindered tumorigenesis in vivo. Moreover, mechanistic investigations including RNA immunoprecipitation and ChIP assays validated that AFAP1-AS1 repressed HMG box-containing protein 1 (HBP1) expression by recruiting LSD1 to the HBP1 promoter regions in PC-9 and H1975 cells. Furthermore, HBP1 functions as a tumor suppressor, and its ectopic expression hindered cell proliferation. Rescue assays determined that the oncogenic effect of AFAP1-AS1 is partially dependent on the epigenetic silencing of HBP1. In conclusion, our results indicate that AFAP1-AS1 is carcinogenic and that the AFAP1-AS1/LSD1/HBP1 axis could constitute a new therapeutic direction for NSCLC.
Collapse
Affiliation(s)
- Shanxun Yu
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Daolu Yang
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yunyao Ye
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Department of OncologyTaizhou People's HospitalTaizhouChina
| | - Pei Liu
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Department of Digestive OncologyThe Fourth Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhenyao Chen
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Tianyao Lei
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jiaze Pu
- Department of Digestive OncologyThe Fourth Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Longfa Liu
- Department of Digestive OncologyThe Fourth Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhaoxia Wang
- Department of OncologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Cancer Medical CenterThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
47
|
Jiang H, Zhu M, Li Y, Li Q. Association between EGFR exon 19 or exon 21 mutations and survival rates after first-line EGFR-TKI treatment in patients with non-small cell lung cancer. Mol Clin Oncol 2019; 11:301-308. [PMID: 31384460 DOI: 10.3892/mco.2019.1881] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/30/2019] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) is the first-line treatment for patients with advanced non-small-cell lung cancer (NSCLC) who have an EGFR mutation. However, little has been reported about the association between EGFR exon 19 deletions or an exon 21 mutation (specifically the L858R point mutation) and survival rates following first-line EGFR-TKI treatment in patients with NSCLC. As a retrospective study, 72 patients with stage IIIB/IV NSCLC carrying EGFR mutations (exon 19 deletions or an exon 21 mutation) were enrolled between 1 January 2008 and 31 December 2013, and all of the patients received first-line EGFR-TKI treatment. The associations between EGFR mutation status or clinical characteristics and response rate (ORR), progression-free survival (PFS) or overall survival (OS) were analyzed. Patients with exon 19 deletions (37 cases) had a higher ORR (75.7 vs. 51.4%; P=0.032), disease control rate (DCR; 89.2 vs. 68.6%; P=0.031), modified median PFS (13.2 vs. 10.8 months; P=0.030) and OS (30.2 vs. 25.6 months; P=0.030) compared with those with an exon 21 mutation (35 cases). Cox multivariate analysis indicated that sex, histological type and smoking history were key factors that affected PFS and OS. Mutations status was associated with PFS, but not OS. Following EGFR-TKI therapy, a better ORR, DCR, PFS and OS was observed in patients with EGFR deletions in exon 19 compared with those with an exon 21 mutation. The EGFR mutation status of patients with non-small cell lung cancer may therefore predict the efficacy and prognosis of EGFR-TKI.
Collapse
Affiliation(s)
- Haiying Jiang
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Mei Zhu
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Yanfang Li
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Qian Li
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
48
|
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is a highly malignant disease with a dismal prognosis that is currently being tested for theclinical activity of checkpoint inhibitors. SCLC is associated with smoking and exhibits a high mutational burden. However, low expression of PD-L1 and MHC antigens, as well low levels of immune cell infiltration and rapid tumor progress seems to limit the efficacy of anticancer immunity. Nevertheless, long-term survival was reported from studies using anti-PD-1/PD-L1 and CTLA-4 agents. AREAS COVERED Data of clinical trials of checkpoint inhibitors in SCLC show lower success rates compared to NSCLC. The mechanisms of resistance to immunotherapy are discussed for their relevance to SCLC patients. EXPERT OPINION Although some factors, such as a high mutation rate, favor immunotherapy for SCLC patients, downregulation of MHC class I, low expression of PD-L1, poor tumor infiltration by effector T cells, presence of myeloid-derived suppressor cells as well as regulatory T lymphocytes counteract the immune system activation by checkpoint inhibitors. Furthermore, this tumor develops avascular regions which have immunosuppressive effects and restrict access of lymphocytes and antibodies. In conclusion, immunotherapy in SCLC is effective in highly selected patients with good performance status and special and unknown preconditions contributing to long-lasting responses.
Collapse
Affiliation(s)
- Gerhard Hamilton
- a Department of Surgery , Medical University of Vienna , Vienna , Austria
| | - Barbara Rath
- a Department of Surgery , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
49
|
Bae EA, Seo H, Kim IK, Jeon I, Kang CY. Roles of NKT cells in cancer immunotherapy. Arch Pharm Res 2019; 42:543-548. [DOI: 10.1007/s12272-019-01139-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/28/2019] [Indexed: 01/22/2023]
|
50
|
Filipska M, Pedraz-Valdunciel C, Chaib I, Rosell R. Biological therapies in lung cancer treatment: using our immune system as an ally to defeat the malignancy. Expert Opin Biol Ther 2019; 19:457-467. [PMID: 30763126 DOI: 10.1080/14712598.2019.1582635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Biological therapies, with immunotherapy leading the field, have arisen as one of the quickest expanding areas of research for cancer treatment in the last few years. The clear benefits for patients are undeniable, satisfying the long-awaited necessity of a target-specific therapy. However, its full potential remains still unexploited due to a lack of response in a majority of patients and pending reliable biomarkers. AREAS COVERED This review provides a summarizing view of the current biological therapies for lung cancer, focusing on immunotherapy - including immune checkpoint inhibitors, adoptive cell therapy and vaccines available in clinical/pre-clinical settings or currently in development. A thorough analysis of the technical and functional differences among all therapies is provided, along with a critical discussion of prospective treatments and potential biomarkers. EXPERT OPINION The use of immunotherapy in the treatment of cancer has provided clear benefits for patients. Still, exploitation of the full potential of immune checkpoint inhibitors alone or in combination, or adoptive cell therapies is hampered by, amongst other reasons, the lack of reliable biomarkers and possible adverse immune effects. We postulate that the development of liquid biopsy-based diagnostics will help to overcome these limitations in the near future.
Collapse
Affiliation(s)
- Martyna Filipska
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain
| | - Carlos Pedraz-Valdunciel
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain
| | - Imane Chaib
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain
| | - Rafael Rosell
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain.,b Pangaea Oncology , Laboratory of Molecular Biology , Barcelona, Spain.,c Institute of Oncology Rosell (IOR) , Quiron-Dexeus University Institute , Barcelona , Spain.,d Catalan Institute of Oncology (ICO) , Hospital Germans Trias i Pujol , Badalona , Spain
| |
Collapse
|