1
|
Li X, Xu H, Hong R, Yang H, Xu L, Zheng G, Xie B. Frontline pemetrexed and cisplatin based-chemotherapy combined with NRT promoted the antitumor in a mouse model of lung carcinoma. Int Immunopharmacol 2025; 149:114174. [PMID: 39929101 DOI: 10.1016/j.intimp.2025.114174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
The efficacy of neoantigen-reactive T cells (NRT) therapy in solid tumors, encompassing aspects such as infiltration, recognition, cytotoxicity, and enduring persistence, is notably influenced by the immunological microenvironment. This study endeavors to investigate whether the co-administration of pemetrexed and cisplatin augments the therapeutic efficacy of NRT therapy in lung cancer. Neoantigens were predicted using a comprehensive analysis of mutation data from Lewis lung carcinoma cells and mouse tail tissues. The immunogenicity of NRT cells was assessed through flow cytometry and IFN-γ ELISpot assays. A mouse model of NSCLC was used to investigate the anti-tumor effects of NRT combined with chemotherapy. The combination of NRT cells and chemotherapy significantly inhibited tumor growth in a mouse model, increased CD3+/CD137+ T cells to promote IFN-γ secretion from NRT cells, and up-regulated the levels of inflammatory cytokine proteins including IFN-γ, TNF, IL-6 and IL-10. Immunofluorescence analysis confirmed increased T-cell infiltration in tumor tissues without adverse effects on vital organs. In addition, transcriptome analyses indicated that the tumor microenvironment was altered to favor M1-like macrophages with an increased M1/M2 ratio, creating a pro-inflammatory environment. The integration of NRT with frontline chemotherapy for lung cancer could yield profoundly ideal therapeutic outcomes.
Collapse
Affiliation(s)
- Xiaoqin Li
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Hang Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Rujun Hong
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Haitao Yang
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Lihuan Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Guanying Zheng
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| | - Baosong Xie
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| |
Collapse
|
2
|
Lin ZY, Song Q, Xu K. Drug Delivery System for Cancer Immunotherapy: Potential Roles, Challenge and Recent Advances. Technol Cancer Res Treat 2025; 24:15330338251338390. [PMID: 40270096 PMCID: PMC12035301 DOI: 10.1177/15330338251338390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
Immunotherapy has emerged as a pivotal advancement in oncological therapeutics, representing a paradigm shift from conventional treatment modalities including surgery, radiotherapy, and chemotherapy. This innovative approach demonstrates considerable clinical potential through its capacity to enhance systemic anti-tumor responses via active or passive immunomodulation. Compared to traditional therapies, immunotherapy offers distinct advantages such as broad applicability, rapid therapeutic onset, and reduced adverse effects. However, critical challenges persist in clinical implementation, particularly concerning treatment safety and efficacy optimization. Current limitations, including drug off-target effects and biological delivery barriers, frequently result in suboptimal therapeutic outcomes and severe complications such as autoimmune disorders and nonspecific inflammation. Recently advancements in drug delivery systems (DDS) present transformative solutions to these challenges. Sophisticated DDS platforms enable precise spatiotemporal delivery of tumor antigens, immunotherapeutic agents, and immunostimulatory molecules, thereby achieving targeted modulation of diverse immune cell populations. This technological innovation not only enhances therapeutic efficacy but also significantly mitigates adverse reactions, while facilitating synergistic combinations with conventional cancer treatments. In this review, we outline the application of new drug delivery platforms in major malignancies (including but not limited to melanoma, non-small cell lung cancer, hormone receptor-positive breast cancer, and hepatocellular carcinoma). We further propose evidence-based optimization strategies for next-generation delivery platforms, aiming to bridge the gap between preclinical development and clinical implementation in cancer immunotherapy.
Collapse
Affiliation(s)
- Zi-Yue Lin
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qian Song
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kai Xu
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Kole E, Jadhav K, Singh R, Mandpe S, Abhang A, Verma RK, Naik J. Recent Developments in Tyrosine Kinase Inhibitor-based Nanotherapeutics for EGFR-resistant Non-small Cell Lung Cancer. Curr Drug Deliv 2025; 22:249-260. [PMID: 38275043 DOI: 10.2174/0115672018278617231207051907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/18/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
The advent of drug resistance in response to epidermal growth factor receptor (EGFR)- tyrosine kinase inhibitor (TKI) targeted therapy represents a serious challenge in the management of non-small cell lung cancer (NSCLC). These acquired resistance mutations, attributed to several advanced EGFR mutations and, necessitated the development of new-generation TKIs. Nanomedicine approaches provide a plausible way to address these problems by providing targeted delivery and sustained release, which have demonstrated success in preclinical trials. This review article provides a summary of nano-formulations designed for EGFR-TKI-resistant NSCLC, highlighting their efficacy in both in vitro and in vivo models. These findings reveal insights into the design of nanoparticles and multifunctional nanosystems, offering a potential avenue for efficacious treatment of EGFR-TKIresistant NSCLC.
Collapse
Affiliation(s)
- Eknath Kole
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon M.S., 425001, India
| | - Krishna Jadhav
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology, Sahibzada Ajit Singh Nagar (Mohali), Punjab, 140306, India
| | - Raghuraj Singh
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology, Sahibzada Ajit Singh Nagar (Mohali), Punjab, 140306, India
| | - Shilpa Mandpe
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon M.S., 425001, India
| | - Ashwin Abhang
- Department of Biopharmaceutics, Biocon Bristol-Myers Squibb R&D Center (BBRC), Bangalore, India
| | - Rahul K Verma
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology, Sahibzada Ajit Singh Nagar (Mohali), Punjab, 140306, India
| | - Jitendra Naik
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon M.S., 425001, India
| |
Collapse
|
4
|
Zhou Z, Fan B, Qiu Q, Cheng H, Wang L, Wu Y, Xie J, Ni C, Li N. Pan-cancer analysis and experimental validation reveal FAM72D as a potential novel biomarker and therapeutic target in lung adenocarcinoma. Gene 2024; 928:148764. [PMID: 39013484 DOI: 10.1016/j.gene.2024.148764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Cancers, particularly lung adenocarcinoma (LUAD), represent a major global health concern. However, the role of FAM72D in various cancers, including LUAD, remains poorly understood. METHODS We utilized databases such as The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression Project (GTEx) and online tools to investigate the correlation between FAM72D expression and its prognostic, diagnostic, and mutational significance, as well as its impact on immune cell infiltration across multiple cancers. Additionally, we developed LUAD cell lines overexpressing FAM72D to confirm its oncogenic role. RESULTS FAM72D expression was elevated in cancerous tissues compared to noncancerous tissues, with diagnostic and prognostic implications in many cancers, including LUAD. Moreover, associations were identified between FAM72D expression and diverse immune subtypes, alongside factors such as microsatellite instability, neoantigens, and tumour mutational burden across pan-cancers. Additionally, FAM72D was associated with immune infiltration and various immune checkpoint-related genes in LUAD. In vitro experiments demonstrated that FAM72D promoted cell proliferation, colony formation, and migration, while inhibiting apoptosis in LUAD cells. CONCLUSIONS Our study establishes associations between FAM72D expression and diagnosis, prognosis, and tumour immunity across multiple cancers, as well as its oncogenic effects in LUAD. FAM72D shows promise as a biomarker and therapeutic target in LUAD.
Collapse
Affiliation(s)
- Zonglang Zhou
- Department of Respiratory and Critical Care Medicine, Center for Respiratory Medicine, The Fourth Affiliated Hospital, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Bingfu Fan
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Qinming Qiu
- Department of Psychiatry, Huzhou Third Municipal Hospital, Huzhou, China
| | - Hongrong Cheng
- Department of Neurology, The Fourth Affiliated Hospital, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Li Wang
- The Fourth Affiliated Hospital, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yibo Wu
- Department of Orthopedics, Xixi Hospital of Hangzhou, Hangzhou, China
| | - Jun Xie
- Department of Nephrology, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| | - Cheng Ni
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Ning Li
- Department of Respiratory and Critical Care Medicine, Center for Respiratory Medicine, The Fourth Affiliated Hospital, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| |
Collapse
|
5
|
Zhao H, Luo K, Liu M, Cai Y, Liu S, Li S, Zhao Y, Zhang H. Immune regulation and prognostic prediction model establishment and validation of PSMB6 in lung adenocarcinoma. Front Genet 2024; 15:1458047. [PMID: 39507618 PMCID: PMC11538069 DOI: 10.3389/fgene.2024.1458047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Lung cancer is one of the most common malignant tumors, and patients are often diagnosed at an advanced stage, posing a substantial risk to human health, so it is crucial to establish a model to forecast the prognosis of patients with lung cancer. Recent research has indicated that proteasome 20S subunit 6 (PSMB6) may be closely associated with anti-apoptotic pathways, and proliferation transduction signals in tumor cells of different tumors. However, the precise role of PSMB6 in the immunoregulatory processes within lung adenocarcinoma (LUAD) is yet to be elucidated. By analyzing the TCGA database, we discovered a positive correlation between the expression of PSMB6 and tumor growth trends, and lung adenocarcinoma patients with elevated PSMB6 expression levels had a worse prognosis. Our findings suggest a close correlation between PSMB6 expression levels, immune cell infiltration and immune checkpoint gene expression, which suggests that PSMB6 may become a new independent prognostic indicator. In addition, we developed a prognostic model of PSMB6-regulated immune infiltration-associated genes by analyzing the link between PSMB6 and the immune microenvironment. This model can not only predict the prognosis of lung adenocarcinoma but also forecasts the patient's reaction to immunotherapy. The validity of this research outcome has been confirmed by the GSE31210 and IMvigor210 cohorts. Analysis of the Kaplan-Meier Plotter database indicates that individuals with elevated levels of PSMB6 expression exhibit a poorer prognosis. Additionally, in vitro experiments demonstrated that knockdown of PSMB6 inhibits the proliferation, migration, and invasion of lung adenocarcinoma cells while promoting their apoptosis. Overall, our findings suggest that PSMB6 could remarkably influence the management and treatment of lung adenocarcinoma, opening new avenues for targeted immunotherapeutic strategies.
Collapse
Affiliation(s)
- Haiyang Zhao
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
| | - Kexin Luo
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
| | - Meihan Liu
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuanze Cai
- North Sichuan Medical College, Nanchong, China
| | - Siman Liu
- North Sichuan Medical College, Nanchong, China
| | - Shijuan Li
- Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Yongsheng Zhao
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hongpan Zhang
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Therapeutic Proteins Key Laboratory of Sichuan Province, Nanchong, China
| |
Collapse
|
6
|
Cui S, Yang Y, Lou S, Huang R, Wang J, Chen Z, Xie J. Establish a novel immune-related gene prognostic risk index (IRGPRI) associated with CD8+ cytotoxic T lymphocytes in non-small-cell lung cancer (NSCLC). Heliyon 2024; 10:e38324. [PMID: 39397989 PMCID: PMC11466668 DOI: 10.1016/j.heliyon.2024.e38324] [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: 04/22/2024] [Revised: 09/22/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
Background The aim of this study is to create an index called IRGPRI (immune-related gene prognostic risk index) that can be utilized for predicting the prognosis and assessing the efficacy of immune checkpoint inhibitors (ICIs) therapy in patients with non-small-cell lung cancer (NSCLC). Methods Distinguishing gene expression patterns (DEGs) were detected in CD8+ cytotoxic T lymphocytes (CTLs) compared to other cellular types such as CD4 T cells, B cells, plasma cells, and CD8 Tex using the advanced technology of Single-cell RNA Sequencing (scRNA-seq). The construction of IRGPRI was accomplished by employing LASSO Cox regression analysis. We conducted a comparative analysis on clinical characteristics and molecular features, such as pathway enrichment and gene mutation, among the distinct subgroups of IRGPRI. Furthermore, we explored the correlation between immunological characteristics and IRGPRI subgroups to comprehensively assess the effectiveness of ICIs in NSCLC patients. Results A total of 109 genes were identified by intersecting immune-related genes with DEGs obtained from single-cell RNA sequencing data (GSE131907), specifically comparing CTLs to other cell types. From these, we selected 7 prognosis-related genes, namely TRBC1, HLA-DMA, CTSH, RAC1, CTSL, ANXA2, and CEBPB. These genes were used to construct the IRGPRI. The prognosis of patients diagnosed with NSCLC was found to be significantly better in the low-risk group compared to the high-risk group, as demonstrated by Kaplan-Meier (K-M) survival analysis. This observation was further confirmed through the utilization of data from the GEO cohort. The low-risk group demonstrated an increase in pathways linked with immune response, whereas the high-risk group exhibited a higher prevalence of pathways related to cancer. Furthermore, it was noted in the TCGA cohort that there existed a significant rise in the mutation frequency of every gene within the high-risk group as opposed to the low-risk group. Missense variation emerged as the most prevalent form of mutation. According to the analysis of immune cell infiltration and function, the comprehensive findings suggest that the group with a low risk is characterized by an increased presence of plasma cells, CTLs, T cells follicular helper, Tregs, and Dendritic cell resting. Additionally, they exhibit a higher score in terms of immune function for B cells, CD8+ T cells, checkpoint activity, T cell inhibition and stimulation. Moreover, this low-risk group demonstrates greater efficacy when treated with ICIs therapy compared to the high-risk group. Conclusions Our research effectively developed and verified a unique IRGPRI, showcasing its association with immune-related characteristics. As a result, the potential of IRGPRI as a valuable biomarker for predicting prognosis and evaluating the effectiveness of ICIs treatment in cancer is evident.
Collapse
Affiliation(s)
- Shenjing Cui
- Department of Clinical Laboratory, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yikun Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Shuang Lou
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Rong Huang
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jing Wang
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhongbiao Chen
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jingjing Xie
- Department of Medical Administration, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
7
|
Wang Y, Chen R, Guo Z, Wei W, Wang T, Ouyang R, Yuan X, Xing Y, Wang F, Wu S, Hou H. Immunological profiling for short-term predictive analysis in PD-1/PD-L1 therapy for lung cancer. BMC Cancer 2024; 24:851. [PMID: 39026211 PMCID: PMC11256628 DOI: 10.1186/s12885-024-12628-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors, such as anti-programmed cell death-1 (PD-1) and PD-1 ligand-1 (PD-L1) antibodies, have achieved breakthrough results in improving long-term survival rates in lung cancer. Although high levels of PD-L1 expression and tumor mutational burden have emerged as pivotal biomarkers, not all patients derive lasting benefits, and resistance to immune checkpoint blockade remains a prevalent issue. Comprehending the immunological intricacies of lung cancer is crucial for uncovering the mechanisms that govern responses and resistance to immunomodulatory treatments. This study aimed to explore the potential of peripheral immune markers in predicting treatment efficiency among lung cancer patients undergoing PD-1/PD-L1 checkpoint inhibitors. METHODS This study enrolled 71 lung cancer patients undergoing PD-1/PD-L1 inhibitor therapy and 20 healthy controls. Immune cell subsets (CD4 + T cells, CD8 + T cells, B cells, NK cells, and NKT cells), phenotypic analysis of T cells and B cells, and PMA/Ionomycin-stimulated lymphocyte function assay were conducted. RESULTS Lung cancer patients exhibited significant alterations in immune cell subsets, notably an increased percentage of Treg cells. Post-treatment, there were substantial increases in absolute numbers of CD3 + T cells, CD8 + T cells, and NKT cells, along with heightened HLA-DR expression on CD3 + T and CD8 + T cells. Comparison between complete remission and non-complete remission (NCR) groups showed higher Treg cell percentages and HLA-DR + CD4 + T cells in the NCR group. CONCLUSION The study findings suggest potential predictive roles for immune cell subsets and phenotypes, particularly Treg cells, HLA-DR + CD4 + T cells, and naïve CD4 + T cells, in evaluating short-term PD-1/PD-L1 therapy efficacy for lung cancer patients. These insights offer valuable prospects for personalized treatment strategies and underscore the importance of immune profiling in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Yun Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Rujia Chen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Zhenzhou Guo
- Department of Laboratory Medicine, Xinfeng County People's Hospital, Ganzhou, China
| | - Wei Wei
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Ting Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Renren Ouyang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Xu Yuan
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Yutong Xing
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China.
| | - Shiji Wu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China.
| | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 1095, 430030, 430030, China.
| |
Collapse
|
8
|
Xiang Y, Liu X, Wang Y, Zheng D, Meng Q, Jiang L, Yang S, Zhang S, Zhang X, Liu Y, Wang B. Mechanisms of resistance to targeted therapy and immunotherapy in non-small cell lung cancer: promising strategies to overcoming challenges. Front Immunol 2024; 15:1366260. [PMID: 38655260 PMCID: PMC11035781 DOI: 10.3389/fimmu.2024.1366260] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/18/2024] [Indexed: 04/26/2024] Open
Abstract
Resistance to targeted therapy and immunotherapy in non-small cell lung cancer (NSCLC) is a significant challenge in the treatment of this disease. The mechanisms of resistance are multifactorial and include molecular target alterations and activation of alternative pathways, tumor heterogeneity and tumor microenvironment change, immune evasion, and immunosuppression. Promising strategies for overcoming resistance include the development of combination therapies, understanding the resistance mechanisms to better use novel drug targets, the identification of biomarkers, the modulation of the tumor microenvironment and so on. Ongoing research into the mechanisms of resistance and the development of new therapeutic approaches hold great promise for improving outcomes for patients with NSCLC. Here, we summarize diverse mechanisms driving resistance to targeted therapy and immunotherapy in NSCLC and the latest potential and promising strategies to overcome the resistance to help patients who suffer from NSCLC.
Collapse
Affiliation(s)
- Yuchu Xiang
- West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Xudong Liu
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yifan Wang
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Dawei Zheng
- The College of Life Science, Sichuan University, Chengdu, China
| | - Qiuxing Meng
- Department of Laboratory Medicine, Liuzhou People’s Hospital, Liuzhou, China
- Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People’s Hospital), Liuzhou, China
| | - Lingling Jiang
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Sha Yang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, China
| | - Sijia Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhang
- Zhongshan Hospital of Fudan University, Xiamen, Fujian, China
| | - Yan Liu
- Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Bo Wang
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
9
|
Wu S, Sheng Q, Liu P, Jiao Z, Lv J, Qiao R, Xie D, Wang Z, Ge J, Li P, Wei T, Lei J, Fan J, Wang L. M1 macrophage-related gene model for NSCLC immunotherapy response prediction. Acta Biochim Biophys Sin (Shanghai) 2024; 56:379-392. [PMID: 38379417 PMCID: PMC10984861 DOI: 10.3724/abbs.2023262] [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: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 02/22/2024] Open
Abstract
Patients diagnosed with non-small cell lung cancer (NSCLC) have a limited lifespan and exhibit poor immunotherapy outcomes. M1 macrophages have been found to be essential for antitumor immunity. This study aims to develop an immunotherapy response evaluation model for NSCLC patients based on transcription. RNA sequencing profiles of 254 advanced-stage NSCLC patients treated with immunotherapy are downloaded from the POPLAR and OAK projects. Immune cell infiltration in NSCLC patients is examined, and thereafter, different coexpressed genes are identified. Next, the impact of M1 macrophage-related genes on the prognosis of NSCLC patients is investigated. Six M1 macrophage coexpressed genes, namely, NKX2-1, CD8A , SFTA3, IL2RB, IDO1, and CXCL9, exhibit a strong association with the prognosis of NSCLC and serve as effective predictors for immunotherapy response. A response model is constructed using a Cox regression model and Lasso Cox regression analysis. The M1 genes are validated in our TD-FOREKNOW NSCLC clinical trial by RT-qPCR. The response model shows excellent immunotherapy response prediction and prognosis evaluation value in advanced-stage NSCLC. This model can effectively predict advanced NSCLC prognosis and aid in identifying patients who could benefit from customized immunotherapy as well as sensitive drugs.
Collapse
Affiliation(s)
- Sifan Wu
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Qiqi Sheng
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
- Department of Thoracic Surgerythe Second Affiliated Hospital of Air Force Medical UniversityXi’an710038China
| | - Pengjun Liu
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Zhe Jiao
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
- Department of Thoracic Surgerythe Second Affiliated Hospital of Air Force Medical UniversityXi’an710038China
| | - Jinru Lv
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
- Department of Thoracic Surgerythe Second Affiliated Hospital of Air Force Medical UniversityXi’an710038China
| | - Rong Qiao
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Dongkun Xie
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Zanhan Wang
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Jiamei Ge
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Penghui Li
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Tiaoxia Wei
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| | - Jie Lei
- Department of Thoracic Surgerythe Second Affiliated Hospital of Air Force Medical UniversityXi’an710038China
| | - Jieyi Fan
- Department of Aerospace MedicineFourth Military Medical UniversityXi’an710032China
| | - Liang Wang
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi’an710032China
| |
Collapse
|
10
|
Yang Y, Xin D, Guan L, Luo X, Wu H, Chu J, Xing J, Liu C, Wang F. Dual immunotherapy in advanced or metastatic non-small cell lung cancer: A network meta-analysis. Heliyon 2024; 10:e27576. [PMID: 38463838 PMCID: PMC10923855 DOI: 10.1016/j.heliyon.2024.e27576] [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: 11/24/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024] Open
Abstract
Objectives Recently, there has been extensive research on dual immunotherapy for advanced or metastatic non-small cell lung cancer (NSCLC), yet a comprehensive evaluation is lacking. This study aimed to rank the available treatment options and assess the efficacy and safety of dual immunotherapy regimens through the implementation of a Bayesian network meta-analysis (NMA). Materials and methods A thorough search was conducted to recognize eligible randomized controlled trials (RCTs) on March 20, 2023. Overall survival (OS), progression-free survival (PFS), treatment-related adverse events (TRAEs) and grade ≥3 TRAEs were evaluated to identify the efficacy and safety of dual immunotherapy regimens. The surface under the cumulative ranking curve (SUCRA) and P score were employed to rank the treatments. Results Eleven clinical trials involving six different regimens were included in this study. The combination of anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) antibodies with anti-T-cell immunoglobulin and ITIM domain (TIGIT) antibodies emerged as the most promising regimen for improving OS and PFS, followed by anti-PD-1/PD-L1 + anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) + chemotherapy treatment and anti-PD-1/PD-L1 + anti-CTLA-4 treatment. The forest plots demonstrated that these three regimens were all superior to chemotherapy. The above results were observed in both unselected treatment line and first-line settings. The least likely to be associated with TRAEs and grade ≥3 TRAEs were respectively anti-CTLA-4 treatment and anti-PD-1/PD-L1 + anti-TIGIT treatment, with anti-PD-1/PD-L1 + anti-CTLA-4 + chemotherapy treatment to be the worst. Conclusions This NMA validated the promising efficacy and safety of dual immunotherapy in advanced or metastatic NSCLC. Among them, anti-PD-1/PD-L1 + anti-TIGIT regimen emerges as a highly potential therapeutic approach. Ongoing research efforts should focus on improving treatment regimens, identifying biomarkers, and managing TRAEs to optimize the patient benefits of dual immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dao Xin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lulu Guan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xi Luo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Han Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jingwen Chu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jianxiang Xing
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chengjiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, Anqing 246000, China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
11
|
Jin K, Li Y, Wei R, Liu Y, Wang S, Tian H. BZW2 promotes malignant progression in lung adenocarcinoma through enhancing the ubiquitination and degradation of GSK3β. Cell Death Discov 2024; 10:105. [PMID: 38424042 PMCID: PMC10904796 DOI: 10.1038/s41420-024-01879-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024] Open
Abstract
The role of Basic leucine zipper and W2 domains 2 (BZW2) in the advancement of different types of tumors is noteworthy, but its involvement and molecular mechanisms in lung adenocarcinoma (LUAD) remain uncertain. Through this investigation, it was found that the upregulation of BZW2 was observed in LUAD tissues, which was associated with an unfavorable prognosis for individuals diagnosed with LUAD, as indicated by data from Gene Expression Omnibus and The Cancer Genome Atlas databases. Based on the clinicopathologic characteristics of LUAD patients from the tissue microarray, both univariate and multivariate analyses indicated that BZW2 functioned as an independent prognostic factor for LUAD. In terms of mechanism, BZW2 interacted with glycogen synthase kinase-3 beta (GSK3β) and enhanced the ubiquitination-mediated degradation of GSK3β through slowing down of the dissociation of the ubiquitin ligase complex, which consists of GSK3β and TNF receptor-associated factor 6. Moreover, BZW2 stimulated Wnt/β-catenin signaling pathway through GSK3β, thereby facilitating the advancement of LUAD. In conclusion, BZW2 was a significant promoter of LUAD. The research we conducted identified a promising diagnostic and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Kai Jin
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yongmeng Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Ruyuan Wei
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yanfei Liu
- Department of Anesthesiology, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Shuai Wang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Lahiri A, Maji A, Potdar PD, Singh N, Parikh P, Bisht B, Mukherjee A, Paul MK. Lung cancer immunotherapy: progress, pitfalls, and promises. Mol Cancer 2023; 22:40. [PMID: 36810079 PMCID: PMC9942077 DOI: 10.1186/s12943-023-01740-y] [Citation(s) in RCA: 472] [Impact Index Per Article: 236.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/22/2022] [Indexed: 02/23/2023] Open
Abstract
Lung cancer is the primary cause of mortality in the United States and around the globe. Therapeutic options for lung cancer treatment include surgery, radiation therapy, chemotherapy, and targeted drug therapy. Medical management is often associated with the development of treatment resistance leading to relapse. Immunotherapy is profoundly altering the approach to cancer treatment owing to its tolerable safety profile, sustained therapeutic response due to immunological memory generation, and effectiveness across a broad patient population. Different tumor-specific vaccination strategies are gaining ground in the treatment of lung cancer. Recent advances in adoptive cell therapy (CAR T, TCR, TIL), the associated clinical trials on lung cancer, and associated hurdles are discussed in this review. Recent trials on lung cancer patients (without a targetable oncogenic driver alteration) reveal significant and sustained responses when treated with programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) checkpoint blockade immunotherapies. Accumulating evidence indicates that a loss of effective anti-tumor immunity is associated with lung tumor evolution. Therapeutic cancer vaccines combined with immune checkpoint inhibitors (ICI) can achieve better therapeutic effects. To this end, the present article encompasses a detailed overview of the recent developments in the immunotherapeutic landscape in targeting small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). Additionally, the review also explores the implication of nanomedicine in lung cancer immunotherapy as well as the combinatorial application of traditional therapy along with immunotherapy regimens. Finally, ongoing clinical trials, significant obstacles, and the future outlook of this treatment strategy are also highlighted to boost further research in the field.
Collapse
Affiliation(s)
- Aritraa Lahiri
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Avik Maji
- Department of Radiation Oncology, N. R. S. Medical College & Hospital, 138 A.J.C. Bose Road, Kolkata, 700014, India
| | - Pravin D Potdar
- Department of Molecular Medicine and Stem Cell Biology, Jaslok Hospital and Research Centre, Mumbai, 400026, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Purvish Parikh
- Department of Clinical Hematology, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, 302022, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, 400012, India
| | - Bharti Bisht
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Anubhab Mukherjee
- Esperer Onco Nutrition Pvt Ltd, 4BA, 4Th Floor, B Wing, Gundecha Onclave, Khairani Road, Sakinaka, Andheri East, Mumbai, Maharashtra, 400072, India.
| | - Manash K Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
13
|
Rahman MM, Masum MHU, Talukder A, Akter R. An in silico reverse vaccinology approach to design a novel multiepitope peptide vaccine for non-small cell lung cancers. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
14
|
Berner F, Bomze D, Lichtensteiger C, Walter V, Niederer R, Hasan Ali O, Wyss N, Bauer J, Freudenmann LK, Marcu A, Wolfschmitt EM, Haen S, Gross T, Abdou MT, Diem S, Knöpfli S, Sinnberg T, Hofmeister K, Cheng HW, Toma M, Klümper N, Purde MT, Pop OT, Jochum AK, Pascolo S, Joerger M, Früh M, Jochum W, Rammensee HG, Läubli H, Hölzel M, Neefjes J, Walz J, Flatz L. Autoreactive napsin A-specific T cells are enriched in lung tumors and inflammatory lung lesions during immune checkpoint blockade. Sci Immunol 2022; 7:eabn9644. [PMID: 36054337 DOI: 10.1126/sciimmunol.abn9644] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cancer treatment with immune checkpoint blockade (ICB) often induces immune-related adverse events (irAEs). We hypothesized that proteins coexpressed in tumors and normal cells could be antigenic targets in irAEs and herein described DITAS (discovery of tumor-associated self-antigens) for their identification. DITAS computed transcriptional similarity between lung tumors and healthy lung tissue based on single-sample gene set enrichment analysis. This identified 10 lung tissue-specific genes highly expressed in the lung tumors. Computational analysis was combined with functional T cell assays and single-cell RNA sequencing of the antigen-specific T cells to validate the lung tumor self-antigens. In patients with non-small cell lung cancer (NSCLC) treated with ICB, napsin A was a self-antigen that elicited strong CD8+ T cell responses, with ICB responders harboring higher frequencies of these CD8+ T cells compared with nonresponders. Human leukocyte antigen (HLA) class I ligands derived from napsin A were present in human lung tumors and in nontumor lung tissues, and napsin A tetramers confirmed the presence of napsin A-specific CD8+ T cells in blood and tumors of patients with NSCLC. Napsin A-specific T cell clonotypes were enriched in lung tumors and ICB-induced inflammatory lung lesions and could kill immortalized HLA-matched NSCLC cells ex vivo. Single-cell RNA sequencing revealed that these T cell clonotypes expressed proinflammatory cytokines and cytotoxic markers. Thus, DITAS successfully identified self-antigens, including napsin A, that likely mediate effective antitumor T cell responses in NSCLC and may simultaneously underpin lung irAEs.
Collapse
Affiliation(s)
- Fiamma Berner
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - David Bomze
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Vincent Walter
- Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Rebekka Niederer
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Omar Hasan Ali
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Nina Wyss
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Jens Bauer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Lena Katharina Freudenmann
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Ana Marcu
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Eva-Maria Wolfschmitt
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Sebastian Haen
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Thorben Gross
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Marie-Therese Abdou
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Stefan Diem
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Stella Knöpfli
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Tobias Sinnberg
- Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Kathrin Hofmeister
- Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Marieta Toma
- Institute of Pathology, University Hospital Bonn (UKB), University of Bonn, Bonn, Germany
| | - Niklas Klümper
- Department of Urology, University Hospital Bonn (UKB), University of Bonn, Bonn, Germany.,Institute of Experimental Oncology, University Hospital Bonn (UKB), University of Bonn, Bonn, Germany
| | - Mette-Triin Purde
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Oltin Tiberiu Pop
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Ann-Kristin Jochum
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Steve Pascolo
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Markus Joerger
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Martin Früh
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Oncology, University of Bern, Bern, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hans-Georg Rammensee
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Heinz Läubli
- Division of Oncology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn (UKB), University of Bonn, Bonn, Germany
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Netherlands
| | - Juliane Walz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany.,Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and Robert Bosch Center for Tumor Diseases (RBCT), Stuttgart, Germany
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany.,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
15
|
Berner F, Niederer R, Luimstra JJ, Pop OT, Jochum AK, Purde MT, Hasan Ali O, Bomze D, Bauer J, Freudenmann LK, Marcu A, Wolfschmitt EM, Haen S, Gross T, Dubbelaar ML, Abdou MT, Baumgaertner P, Appenzeller C, Cicin-Sain C, Lenz T, Speiser DE, Ludewig B, Driessen C, Jörger M, Früh M, Jochum W, Cozzio A, Rammensee HG, Walz J, Neefjes J, Flatz L. Keratinocyte differentiation antigen-specific T cells in immune checkpoint inhibitor-treated NSCLC patients are associated with improved survival. Oncoimmunology 2021; 10:2006893. [PMID: 34858733 PMCID: PMC8632109 DOI: 10.1080/2162402x.2021.2006893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have improved the survival of patients with non-small cell lung cancer (NSCLC) by reinvigorating tumor-specific T cell responses. However, the specificity of such T cells and the human leukocyte antigen (HLA)-associated epitopes recognized, remain elusive. In this study, we identified NSCLC T cell epitopes of recently described NSCLC-associated antigens, termed keratinocyte differentiation antigens. Epitopes of these antigens were presented by HLA-A 03:01 and HLA-C 04:01 and were associated with responses to ICI therapy. Patients with CD8+ T cell responses to these epitopes had improved overall and progression-free survival. T cells specific for such epitopes could eliminate HLA class I-matched NSCLC cells ex vivo and were enriched in patient lung tumors. The identification of novel lung cancer HLA-associated epitopes that correlate with improved ICI-dependent treatment outcomes suggests that keratinocyte-specific proteins are important tumor-associated antigens in NSCLC. These findings improve our understanding of the mechanisms of ICI therapy and may help support the development of vaccination strategies to improve ICI-based treatment of these tumors.
Collapse
Affiliation(s)
- Fiamma Berner
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Rebekka Niederer
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Jolien J Luimstra
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Oltin Tiberiu Pop
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Ann-Kristin Jochum
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Mette-Triin Purde
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Omar Hasan Ali
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada.,Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - David Bomze
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jens Bauer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Lena Katharina Freudenmann
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Ana Marcu
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Eva-Maria Wolfschmitt
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Sebastian Haen
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Thorben Gross
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Marissa Lisa Dubbelaar
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Marie-Therese Abdou
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Petra Baumgaertner
- Department of Oncology, Ludwig Cancer Research, University of Lausanne, Switzerland
| | - Christina Appenzeller
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Caroline Cicin-Sain
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Tobias Lenz
- Research Unit Evolutionary Immunogenomics, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Daniel E Speiser
- Department of Oncology, Ludwig Cancer Research, University of Lausanne, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Christoph Driessen
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Markus Jörger
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Martin Früh
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Oncology, University of Bern, Bern, Switzerland
| | - Wolfram Jochum
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Antonio Cozzio
- Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hans-Georg Rammensee
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Juliane Walz
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany.,Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and Robert Bosch Center for Tumor Diseases (RBCT), Stuttgart, Germany
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
FKBP10 Regulates Protein Translation to Sustain Lung Cancer Growth. Cell Rep 2021; 30:3851-3863.e6. [PMID: 32187554 DOI: 10.1016/j.celrep.2020.02.082] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 10/29/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer therapy is limited, in part, by lack of specificity. Thus, identifying molecules that are selectively expressed by, and relevant for, cancer cells is of paramount medical importance. Here, we show that peptidyl-prolyl-cis-trans-isomerase (PPIase) FK506-binding protein 10 (FKBP10)-positive cells are present in cancer lesions but absent in the healthy parenchyma of human lung. FKBP10 expression negatively correlates with survival of lung cancer patients, and its downregulation causes a dramatic diminution of lung tumor burden in mice. Mechanistically, our results from gain- and loss-of-function assays show that FKBP10 boosts cancer growth and stemness via its PPIase activity. Also, FKBP10 interacts with ribosomes, and its downregulation leads to reduction of translation elongation at the beginning of open reading frames (ORFs), particularly upon insertion of proline residues. Thus, our data unveil FKBP10 as a cancer-selective molecule with a key role in translational reprogramming, stem-like traits, and growth of lung cancer.
Collapse
|
17
|
Tubin S, Khan MK, Gupta S, Jeremic B. Biology of NSCLC: Interplay between Cancer Cells, Radiation and Tumor Immune Microenvironment. Cancers (Basel) 2021; 13:775. [PMID: 33673332 PMCID: PMC7918834 DOI: 10.3390/cancers13040775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
The overall prognosis and survival of non-small cell lung cancer (NSCLC) patients remain poor. The immune system plays an integral role in driving tumor control, tumor progression, and overall survival of NSCLC patients. While the tumor cells possess many ways to escape the immune system, conventional radiotherapy (RT) approaches, which are directly cytotoxic to tumors, can further add additional immune suppression to the tumor microenvironment by destroying many of the lymphocytes that circulate within the irradiated tumor environment. Thus, the current immunogenic balance, determined by the tumor- and radiation-inhibitory effects is significantly shifted towards immunosuppression, leading to poor clinical outcomes. However, newer emerging evidence suggests that tumor immunosuppression is an "elastic process" that can be manipulated and converted back into an immunostimulant environment that can actually improve patient outcome. In this review we will discuss the natural immunosuppressive effects of NSCLC cells and conventional RT approaches, and then shift the focus on immunomodulation through novel, emerging immuno- and RT approaches that promise to generate immunostimulatory effects to enhance tumor control and patient outcome. We further describe some of the mechanisms by which these newer approaches are thought to be working and set the stage for future trials and additional preclinical work.
Collapse
Affiliation(s)
- Slavisa Tubin
- MedAustron Ion Therapy Center, Marie Curie-Straße 5, 2700 Wiener Neustadt, Austria
| | - Mohammad K. Khan
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA;
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Branislav Jeremic
- Research Institute of Clinical Medicine, 13 Tevdore Mgdveli, Tbilisi 0112, Georgia;
| |
Collapse
|
18
|
PD-L1 promotes tumor growth and progression by activating WIP and β-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis 2020; 11:506. [PMID: 32632098 PMCID: PMC7338457 DOI: 10.1038/s41419-020-2701-z] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/10/2020] [Accepted: 06/14/2020] [Indexed: 12/13/2022]
Abstract
PD-L1 is overexpressed in tumor cells and contributes to cancer immunoevasion. However, the role of the tumor cell-intrinsic PD-L1 in cancers remains unknown. Here we show that PD-L1 regulates lung cancer growth and progression by targeting the WIP and β-catenin signaling. Overexpression of PD-L1 promotes tumor cell growth, migration and invasion in lung cancer cells, whereas PD-L1 knockdown has the opposite effects. We have also identified WIP as a new downstream target of PD-L1 in lung cancer. PD-L1 positively modulates the expression of WIP. Knockdown of WIP also inhibits cell viability and colony formation, whereas PD-L1 overexpression can reverse this inhibition effects. In addition, PD-L1 can upregulate β-catenin by inhibiting its degradation through PI3K/Akt signaling pathway. Moreover, we show that in lung cancer cells β-catenin can bind to the WIP promoter and activate its transcription, which can be promoted by PD-L1 overexpression. The in vivo experiments in a human lung cancer mouse model have also confirmed the PD-L1-mediated promotion of tumor growth and progression through activating the WIP and β-catenin pathways. Furthermore, we demonstrate that PD-L1 expression is positively correlated with WIP in tumor tissues of human adenocarcinoma patients and the high expression of PD-L1 and WIP predicts poor prognosis. Collectively, our results provide new insights into understanding the pro-tumorigenic role of PD-L1 and its regulatory mechanism on WIP in lung cancer, and suggest that the PD-L1/Akt/β-catenin/WIP signaling axis may be a potential therapeutic target for lung cancers.
Collapse
|
19
|
Lema DA, Jankowska-Gan E, Sethakorn N, Burlingham W, Leal T. Identification of PD1-mediated regulation of antitumor antigen response in patients with NSCLC using the trans vivo DTH assay. J Immunother Cancer 2020; 8:jitc-2019-000152. [PMID: 32527929 PMCID: PMC7292037 DOI: 10.1136/jitc-2019-000152] [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] [Accepted: 05/07/2020] [Indexed: 11/24/2022] Open
Abstract
Objectives Emerging evidence has shown a role for tumor antigen-specific regulation in cancer. Identifying individuals with pre-existing regulatory responses may be key to understand those who are more likely to respond to Programmed Death-1 (PD-1) or PD-1 Ligand 1 (PD-L1) checkpoint blockade. We hypothesized that a functional assay could identify the role of PD-1/PD-L1 interactions on tumor-specific immune cells in the peripheral blood in patients with advanced non-small-cell lung cancer (NSCLC). Methods We performed the trans vivo delayed-type hypersensitivity assay to identify the role of PD-1/PD-L1-mediated tumor-specific immune regulation in ten patients with advanced NSCLC. Results The majority of patients had PD-1-mediated anergic immune responses towards their tumor antigens. Eight out of nine of these patients did not respond to their own tumor antigens but responded in the presence of anti-PD-1 antibody (‘PD-1 anergy’ phenotype). A minority (3/9) also had ‘active’ PD-1-mediated immune suppressive regulatory responses. Our results suggest that PD-1-anergy is a common feature of NSCLC immune responses, whereas PD-1-mediated immune suppression is present only in a minority of patients. The latter was associated with poor clinical outcomes in our sample. Conclusions Overall, our results indicate that bystander suppression or the ‘anergy-only’ phenomenon may be novel biomarkers in NSCLC and suggest prediction value based on these phenotypes.
Collapse
Affiliation(s)
- Diego A Lema
- Surgery-Transplant, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Ewa Jankowska-Gan
- Surgery-Transplant, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Nan Sethakorn
- Medicine, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - William Burlingham
- Surgery-Transplant, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Ticiana Leal
- Medicine, University of Wisconsin Madison, Madison, Wisconsin, USA
| |
Collapse
|
20
|
Grimaldi A, Cammarata I, Martire C, Focaccetti C, Piconese S, Buccilli M, Mancone C, Buzzacchino F, Berrios JRG, D'Alessandris N, Tomao S, Giangaspero F, Paroli M, Caccavale R, Spinelli GP, Girelli G, Peruzzi G, Nisticò P, Spada S, Panetta M, Letizia Cecere F, Visca P, Facciolo F, Longo F, Barnaba V. Combination of chemotherapy and PD-1 blockade induces T cell responses to tumor non-mutated neoantigens. Commun Biol 2020; 3:85. [PMID: 32099064 PMCID: PMC7042341 DOI: 10.1038/s42003-020-0811-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
Here, we developed an unbiased, functional target-discovery platform to identify immunogenic proteins from primary non-small cell lung cancer (NSCLC) cells that had been induced to apoptosis by cisplatin (CDDP) treatment in vitro, as compared with their live counterparts. Among the multitude of proteins identified, some of them were represented as fragmented proteins in apoptotic tumor cells, and acted as non-mutated neoantigens (NM-neoAgs). Indeed, only the fragmented proteins elicited effective multi-specific CD4+ and CD8+ T cell responses, upon a chemotherapy protocol including CDDP. Importantly, these responses further increased upon anti-PD-1 therapy, and correlated with patients' survival and decreased PD-1 expression. Cross-presentation assays showed that NM-neoAgs were unveiled in apoptotic tumor cells as the result of caspase-dependent proteolytic activity of cellular proteins. Our study demonstrates that apoptotic tumor cells generate a repertoire of immunogenic NM-neoAgs that could be potentially used for developing effective T cell-based immunotherapy across multiple cancer patients.
Collapse
MESH Headings
- Aged
- Antigen Presentation/drug effects
- Antigen Presentation/immunology
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/isolation & purification
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Case-Control Studies
- Cell Line, Tumor
- Cisplatin/administration & dosage
- Cisplatin/pharmacology
- Combined Modality Therapy
- Drug Screening Assays, Antitumor/methods
- Female
- Humans
- Immunity, Cellular/drug effects
- Immunotherapy/methods
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Male
- Middle Aged
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/physiology
Collapse
Affiliation(s)
- Alessio Grimaldi
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Ilenia Cammarata
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Carmela Martire
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Chiara Focaccetti
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Silvia Piconese
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Marta Buccilli
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Carmine Mancone
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, 00161, Rome, Italy
| | - Federica Buzzacchino
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo Patologiche, Oncologia Medica, Università di Roma, 00161, Rome, Italy
| | - Julio Rodrigo Giron Berrios
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo Patologiche, Oncologia Medica, Università di Roma, 00161, Rome, Italy
| | - Nicoletta D'Alessandris
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Silverio Tomao
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo Patologiche, Oncologia Medica, Università di Roma, 00161, Rome, Italy
| | - Felice Giangaspero
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Marino Paroli
- Dipartimento di Scienze e Biotecnologie Medico-Chirurgiche, Sapienza Università di Roma - Polo Pontino, 04100, Latina, Italy
| | - Rosalba Caccavale
- Dipartimento di Scienze e Biotecnologie Medico-Chirurgiche, Sapienza Università di Roma - Polo Pontino, 04100, Latina, Italy
| | - Gian Paolo Spinelli
- UOC Oncologia Universitaria, ASL Latina (distretto Aprilia), Sapienza Università di Roma, Via Giustiniano snc, 04011, Aprilia, Latina, Italy
| | - Gabriella Girelli
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, 00161, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Mariangela Panetta
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | | | - Paolo Visca
- Unit of Pathology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Facciolo
- Thoracic Surgery Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Flavia Longo
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo Patologiche, Oncologia Medica, Università di Roma, 00161, Rome, Italy
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy.
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy.
- Istituto Pasteur - Fondazione Cenci Bolognetti, 00185, Rome, Italy.
| |
Collapse
|
21
|
Wu F, Yin Z, Yang L, Fan J, Xu J, Jin Y, Yu J, Zhang D, Yang G. Smoking Induced Extracellular Vesicles Release and Their Distinct Properties in Non-Small Cell Lung Cancer. J Cancer 2019; 10:3435-3443. [PMID: 31293647 PMCID: PMC6603414 DOI: 10.7150/jca.30425] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: Smoking is a strong relative risk factor for lung cancer. Extracellular vesicles (EVs), particularly exosomes, have been implicated in cancers. In this study, we characterized smoking induced extracellular vesicles in smokers with non-small cell lung cancer (NSCLC). Methods: EVs were isolated from bronchoalveolar lavage (BAL) from smokers and NSCLC patients. EV microRNAs (miRNAs) were analyzed by using a TaqMan microRNA assays. Vesicle mRNAs and long non-coding RNAs (lncRNAs) were measured with quantitative RT-PCR. Tumor associated antigens were examined by Western Blot. Results: Higher levels of local site EVs are found in the lung of smokers and NSCLC patients. Further, over 90% of lung EVs are round vesicles of approximately 50-200 nm, ie., exosomes. There are 21 EV miRNAs up regulated, while 10 miRNAs under regulated, in smokers when compared to controls (relative fold > 2, p < 0.05). These miRNAs were further observed to be dysregulated in NSCLC patients when compared to smokers. Bioinformatic analysis demonstrated that Proteoglycans, Fatty acid biosynthesis, ErbB, Hippo, TGF-beta, Wnt, Rap1, AMPK and Ras pathways were the most prominent pathways enriched in NSCLC EV miRNA signatures. In addition, messenger RNA transcripts including EGFR, KRAS, ALK, MET, LKB1, BRAF, PIK3CA, RET, and ROS1 were significantly higher expressed in lung EVs in smokers and NSCLC patients compared to controls. Long non-coding RNAs, including MALAT1, HOTAIR, HOTTIP, AGAP2-AS1, ATB, TCF7, FOXD2-AS1, HOXA11-AS, PCAF1, and BCAR4, were over expressed in EVs from smokers and NSCLC patients. Furthermore, protein levels of tumor associated antigens including BAGE, PD-L1, MAGE-3, and AKAP4 were significantly dysregulated in EVs of smokers and NSCLC patients compared to healthy controls. Conclusions: In conclusion, these data demonstrated an intrinsic relationship of smoking dysregulated EVs and EVs contained RNA, proteins which may involve in the development of NSCLC.
Collapse
Affiliation(s)
- Feng Wu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhongyuan Yin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Yang
- Oncology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinshuo Fan
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Juanjuan Xu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Jin
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jizhang Yu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dan Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guanghai Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
22
|
Carter CA, Zeman K, Day RM, Richard P, Oronsky A, Oronsky N, Lybeck M, Scicinski J, Oronsky B. Addressing the elephant in the room, therapeutic resistance in non-small cell lung cancer, with epigenetic therapies. Oncotarget 2018; 7:40781-40791. [PMID: 27007055 PMCID: PMC5130044 DOI: 10.18632/oncotarget.8205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/07/2016] [Indexed: 12/15/2022] Open
Abstract
Like Chinese boxes nesting inside each other, the classification of non-small cell lung cancer (NSCLC) is subdivided into smaller and smaller subtypes on the basis of histological and molecular attributes. The latter characterizes NSCLC by its molecular alterations and the identification of inhibitors that target these cancer-specific "driver" mutations. Despite the initial promise of precision-guided therapies to inhibit a finer and finer array of molecular subcategories, despite even the curative potential of immunotherapeutic checkpoint blockade, in particular, casualties still abound and true clinical success stories are few and far between; the ever-present, if sometimes unmentioned, "elephant in the room", is the acquisition of resistance, which, sooner or later, rears its ugly head to undermine treatment success and shorten survival. Emerging data suggests that epigenetic therapies are able to reprogram the aberrant tumor-associated epigenome and 'tame the beast of resistance', thereby prolonging survival. This article reviews the role of epigenetic dysregulation in NSCLC, explores PFS2 as a possible surrogate endpoint, briefly mentions possible biomarkers and highlights combinatorial treatment epigenetic strategies to "prime" tumors and reverse resistance.
Collapse
Affiliation(s)
- Corey A Carter
- Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Karen Zeman
- National Naval Medical Center, Bethesda, MD, USA
| | - Regina M Day
- Uniformed Services University of The Health Sciences, Bethesda, MD, USA
| | - Patrick Richard
- Uniformed Services University of The Health Sciences, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
23
|
Cho JH. Immunotherapy for Non-small-cell Lung Cancer: Current Status and Future Obstacles. Immune Netw 2017; 17:378-391. [PMID: 29302251 PMCID: PMC5746608 DOI: 10.4110/in.2017.17.6.378] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is one of the leading causes of death worldwide. There are 2 major subtypes of lung cancer, non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). Studies show that NSCLC is the more prevalent type of lung cancer that accounts for approximately 80%-85% of cases. Although, various treatment methods, such as chemotherapy, surgery, and radiation therapy have been used to treat lung cancer patients, there is an emergent need to develop more effective approaches to deal with advanced stages of tumors. Recently, immunotherapy has emerged as a new approach to combat with such tumors. The development and success of programmed cell death 1 (PD-1)/program death-ligand 1 (PD-L1) inhibitors and cytotoxic T-lymphocyte antigen 4 (CTLA-4) blockades in treating metastatic cancers opens a new pavement for the future research. The current mini review discusses the significance of immune checkpoint inhibitors in promoting the death of tumor cells. Additionally, this review also addresses the importance of tumor-specific antigens (neoantigens) in the development of cancer vaccines and major challenges associated with this therapy. Immunotherapy can be a promising approach to treat NSCLC because it stimulates host's own immune system to recognize cancer cells. Therefore, future research should focus on the development of new methodologies to identify novel checkpoint inhibitors and potential neoantigens.
Collapse
Affiliation(s)
- Ju Hwan Cho
- Arthur G. James Cancer Hospital Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Yi JS, Ready N, Healy P, Dumbauld C, Osborne R, Berry M, Shoemaker D, Clarke J, Crawford J, Tong B, Harpole D, D'Amico TA, McSherry F, Dunphy F, McCall SJ, Christensen JD, Wang X, Weinhold KJ. Immune Activation in Early-Stage Non-Small Cell Lung Cancer Patients Receiving Neoadjuvant Chemotherapy Plus Ipilimumab. Clin Cancer Res 2017; 23:7474-7482. [PMID: 28951518 DOI: 10.1158/1078-0432.ccr-17-2005] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/25/2017] [Accepted: 09/22/2017] [Indexed: 02/04/2023]
Abstract
Purpose: To determine the immunologic effects of neoadjuvant chemotherapy plus ipilimumab in early-stage non-small cell lung cancer (NSCLC) patients.Experimental Design: This is a single-arm chemotherapy plus phased ipilimumab phase II study of 24 treatment-naïve patients with stage IB-IIIA NSCLC. Patients received neoadjuvant therapy consisting of 3 cycles of paclitaxel with either cisplatin or carboplatin and ipilimumab included in the last 2 cycles.Results: Chemotherapy alone had little effect on immune parameters in PBMCs. Profound CD28-dependent activation of both CD4 and CD8 cells was observed following ipilimumab. Significant increases in the frequencies of CD4+ cells expressing activation markers ICOS, HLA-DR, CTLA-4, and PD-1 were apparent. Likewise, increased frequencies of CD8+ cells expressing the same activation markers, with the exception of PD-1, were observed. We also examined 7 resected tumors and found higher frequencies of activated tumor-infiltrating lymphocytes than those observed in PBMCs. Surprisingly, we found 4 cases of preexisting tumor-associated antigens (TAA) responses against survivin, PRAME, or MAGE-A3 present in PBMC at baseline, but neither increased frequencies nor the appearance of newly detectable responses following ipilimumab therapy. Ipilimumab had little effect on the frequencies of circulating regulatory T cells and MDSCs.Conclusions: This study did not meet the primary endpoint of detecting an increase in blood-based TAA T-cell responses after ipilimumab. Collectively, these results highlight the immune activating properties of ipilimumab in early-stage NSCLC. The immune profiling data for ipilimumab alone can contribute to the interpretation of immunologic data from combined immune checkpoint blockade immunotherapies. Clin Cancer Res; 23(24); 7474-82. ©2017 AACR.
Collapse
Affiliation(s)
- John S Yi
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Neal Ready
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Patrick Healy
- Duke Cancer Center Biostatistics, Duke University Medical Center, Durham, North Carolina
| | - Chelsae Dumbauld
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Robyn Osborne
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Mark Berry
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Debra Shoemaker
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jeffrey Clarke
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jeffrey Crawford
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Betty Tong
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - David Harpole
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Thomas A D'Amico
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Frances McSherry
- Duke Cancer Center Biostatistics, Duke University Medical Center, Durham, North Carolina
| | - Frank Dunphy
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Shannon J McCall
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Jared D Christensen
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Xiaofei Wang
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Kent J Weinhold
- Department of Surgery, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
25
|
Zhou Y, Hu Z, Cao S, Yan B, Qian J, Zhong H. Concomitant Mycobacterium tuberculosis infection promotes lung tumor growth through enhancing Treg development. Oncol Rep 2017. [PMID: 28627635 PMCID: PMC5561997 DOI: 10.3892/or.2017.5733] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Lung cancer is the most common malignancy in humans. An increased population of CD4+Foxp3+ regulatory T cells (Tregs) in the tumor-associated microenvironment plays an important role in cancer immune evasion. The exact role and the involved mechanisms of concomitant H37Rv infection in non-small cell lung cancer (NSCLC) development are still not clear. Here, we showed that H37Rv infection promoted NSCLC cell growth with a higher percentage of Tregs found in draining lymph nodes. We also determined in vitro that H37Rv infection induced macrophage maturation and PD-L1 expression, which promoted Treg proportion, with enhanced proliferation suppression function. Mechanism analysis revealed that AKT-mTORC1 signal was important for PD-L1 expression induced by H37Rv infection. Suppressing of AKT-mTORC1 signal by rapamycin or raptor deficiency showed decreased PD-L1 levels which further reduced Treg proportion in a co-culture system. Finally, tumor-bearing mice injected with H37Rv plus rapamycin enhance the immune response of lung cancer compared with injected with H37Rv alone. This study demonstrated that concomitant H37Rv infection promote NSCLC tumor immune eacape through enhancing Treg proportion.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Zhangguo Hu
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Shuhui Cao
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Bo Yan
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Jialin Qian
- Department of Respiration Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Hua Zhong
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
26
|
Jiang L, Jiang S, Lin Y, Yang H, Zhao Z, Xie Z, Lin Y, Long H. Combination of body mass index and oxidized low density lipoprotein receptor 1 in prognosis prediction of patients with squamous non-small cell lung cancer. Oncotarget 2016; 6:22072-80. [PMID: 26061746 PMCID: PMC4673147 DOI: 10.18632/oncotarget.4299] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 05/22/2015] [Indexed: 01/23/2023] Open
Abstract
Lung cancer, especially non-small cell lung cancer (NSCLC), represents enormous challenges in continuously achieving treatment improvements. Besides cancer, obesity is becoming ever more prevalent. Obesity is increasingly acknowledged as a major risk factor for several types of common cancers. Significant mechanisms overlap in the pathobiology of obesity and tumorigenesis. One of these mechanisms involves oxidized low density lipoprotein receptor 1 (OLR1), as a link between obesity and cancer. Additionally, body mass index (BMI) has been widely used in exploiting the role of obesity on a series of diseases, including cancer. Significantly, squamous NSCLC revealed to be divergent clinical and molecular phenotypes compared with non-squamous NSCLC. Consequently, OLR1 immunostaining score and BMI were assessed by Fisher's linear discriminant analysis to discriminate if progression-free survival (PFS) would exceed 2 years. In addition, the final model was utilized to calculate the discriminant score in each study participant. Finally, 131 patients with squamous NCSLC were eligible for analysis. And a prediction model was established for PFS based on these 2 markers and validated in a second set of squamous NCSLC patients. The model offers a novel tool for survival prediction and could establish a framework for future individualized therapy for patients with squamous NCSLC.
Collapse
Affiliation(s)
- Long Jiang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China.,University of California, San Francisco, San Francisco, CA, USA
| | - Shanshan Jiang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yongbin Lin
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Han Yang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zerui Zhao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zehua Xie
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yaobin Lin
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hao Long
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Lung Cancer Institute of Sun Yat-Sen University, Guangzhou, China.,Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
27
|
Mahasa KJ, Ouifki R, Eladdadi A, Pillis LD. Mathematical model of tumor-immune surveillance. J Theor Biol 2016; 404:312-330. [PMID: 27317864 DOI: 10.1016/j.jtbi.2016.06.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/26/2022]
Abstract
We present a novel mathematical model involving various immune cell populations and tumor cell populations. The model describes how tumor cells evolve and survive the brief encounter with the immune system mediated by natural killer (NK) cells and the activated CD8(+) cytotoxic T lymphocytes (CTLs). The model is composed of ordinary differential equations describing the interactions between these important immune lymphocytes and various tumor cell populations. Based on up-to-date knowledge of immune evasion and rational considerations, the model is designed to illustrate how tumors evade both arms of host immunity (i.e. innate and adaptive immunity). The model predicts that (a) an influx of an external source of NK cells might play a crucial role in enhancing NK-cell immune surveillance; (b) the host immune system alone is not fully effective against progression of tumor cells; (c) the development of immunoresistance by tumor cells is inevitable in tumor immune surveillance. Our model also supports the importance of infiltrating NK cells in tumor immune surveillance, which can be enhanced by NK cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Khaphetsi Joseph Mahasa
- DST/NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), University of Stellenbosch, Stellenbosch, South Africa.
| | - Rachid Ouifki
- DST/NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), University of Stellenbosch, Stellenbosch, South Africa
| | | | | |
Collapse
|
28
|
Mi D, Ren W, Yang K. Adoptive immunotherapy with interleukin-2 & induced killer cells in non-small cell lung cancer: A systematic review & meta-analysis. Indian J Med Res 2016; 143:S1-S10. [PMID: 27748271 PMCID: PMC5080917 DOI: 10.4103/0971-5916.191738] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND & OBJECTIVES The effectiveness of interleukin-2 (IL-2) and induced killer cells for non-small cell lung cancer (NSCLC) is controversial. This study evaluates the efficacy and safety of interleukin-2 and induced killer cells on NSCLC, so as to provide references for further clinical practice and research. METHODS Relevant randomized controlled trials (RCTs) were searched in Cochrane library (Issue 2, 2013), Web of Science (1980-March 2013), PubMed (1966-March 2013), China Knowledge Resource Integrated database (CNKI) (1994-March 2013), China Biology Medicine database (CBM) (1978-March 2013), VIP (1989-March 2013), and Wan Fang databases (1997-March 2013). There were no language restrictions. After independent quality assessment and data extraction by two authors, meta-analysis was conducted by RevMan 5.1 software. RESULTS Ten RCTs were included. Odds ratio (OR), 95% confidence intervals (CI), P value expressed as test group (interleukin-2 or induced killer cells combined chemotherapy) versus control group (chemotherapy alone), was 2.02 (1.24, 3.29; P=0.004) for disease control rate. Hazard ratios (HR) (95% CI; P value), expressed as test group (interleukin-2 or induced killer cells) versus control group, were 0.60 (0.46, 0.79; P=0.0003) for overall survival of postoperative treatment, and 0.77 (0.60, 0.99; P =0.04) for overall survival of combination with chemotherapy. Mean differences (MD) (95% CI; P value), expressed as test group (interleukin-2 or induced killer cells) versus control group (after treatment), were 11.32 (6.32, 16.33; P=0.00001) for NK cells, 11.79 (2.71, 20.86; P=0.01) for CD3+ cells, 14.63 (2.62, 26.64; P=0.02) for CD4+ cells, and -4.49 (-7.80, 1.18; P=0.008) for CD8+ cells. INTERPRETATION & CONCLUSIONS The meta-analysis showed that IL-2 or induced killer cells combination therapy was efficacious in treating NSCLC and improved overall survival. Further analysis of trials having adequate information and data need to be done to confirm these findings.
Collapse
Affiliation(s)
- Denghai Mi
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Chengguan District, Lanzhou, Gansu, PR China
- The First Clinical Medicine College of Lanzhou University, 1 Donggang West Road, Chengguan District, Lanzhou, Gansu, PR China
- Gansu Provincial Second People's Hospital, 1 Hezheng West Road, Chengguan District, Lanzhou, Gansu, PR China
| | - Weiwei Ren
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Chengguan District, Lanzhou, Gansu, PR China
- The First Clinical Medicine College of Lanzhou University, 1 Donggang West Road, Chengguan District, Lanzhou, Gansu, PR China
- Gansu Provincial Cancer Hospital, 2 Xiaoxihu East Street, Qilihe District, Lanzhou, Gansu, PR China
| | - Kehu Yang
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Chengguan District, Lanzhou, Gansu, PR China
- The First Clinical Medicine College of Lanzhou University, 1 Donggang West Road, Chengguan District, Lanzhou, Gansu, PR China
| |
Collapse
|
29
|
Jiang L, Zhao Z, Jiang S, Lin Y, Yang H, Xie Z, Lin Y, Long H. Immunological markers predict the prognosis of patients with squamous non-small cell lung cancer. Immunol Res 2016; 62:316-24. [PMID: 25999209 DOI: 10.1007/s12026-015-8662-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lung cancer has become the leading cause of cancer-related death worldwide. However, treatment failures still represent enormous challenges, and it is doubtful whether standard treatment modalities could continuously achieve substantial improvements. As one of the novel therapy strategies, PD-L1 has been shown the function of down-regulating T cell activation through receptor PD-1. Moreover, prognosis of cancer patients is based not only on tumor-related factors but also on host-related factors, particularly systemic inflammatory response. Significantly, squamous non-small cell lung cancer (NSCLC) revealed to be divergent clinical and molecular phenotypes compared with non-squamous NSCLC. Monocyte ratio, neutrophils to lymphocytes ratio, PD-L1 immunostaining score and PD-1-positive stained tumor-infiltrating lymphocyte counts were assessed by Fisher's linear discriminant analysis to discriminate whether overall survival (OS) would exceeding 5 years. Finally, a prediction model was established for OS based on these immunological markers. Furthermore, this prediction model was validated in a second set of squamous NSCLC patients. The model offers a novel tool for survival prediction and could have important clinical implications for patients with squamous NSCLC, thus providing a framework for future individualized therapy.
Collapse
Affiliation(s)
- Long Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651, Dongfeng Rd East, Guangzhou, 510060, People's Republic of China,
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Liu X, Li J, Liu Y, Ding J, Tong Z, Liu Y, Zhou Y, Liu Y. Calreticulin acts as an adjuvant to promote dendritic cell maturation and enhances antigen-specific cytotoxic T lymphocyte responses against non-small cell lung cancer cells. Cell Immunol 2016; 300:46-53. [DOI: 10.1016/j.cellimm.2015.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 01/08/2023]
|
31
|
Liu Y, Tian X, Jiang S, Ren X, Liu F, Yang J, Chen Y, Jiang Y. Umbilical cord blood-derived dendritic cells infected by adenovirus for SP17 expression induce antigen-specific cytotoxic T cells against NSCLC cells. Cell Immunol 2015; 298:18-24. [DOI: 10.1016/j.cellimm.2015.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/09/2015] [Accepted: 08/17/2015] [Indexed: 12/25/2022]
|
32
|
Mirandola L, Figueroa JA, Phan TT, Grizzi F, Kim M, Rahman RL, Jenkins MR, Cobos E, Jumper C, Alalawi R, Chiriva-Internati M. Novel antigens in non-small cell lung cancer: SP17, AKAP4, and PTTG1 are potential immunotherapeutic targets. Oncotarget 2015; 6:2812-2826. [PMID: 25739119 PMCID: PMC4413619 DOI: 10.18632/oncotarget.2802] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/15/2014] [Indexed: 12/03/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths in both genders worldwide, with an incidence only second to prostate cancer in men and breast cancer in women. The lethality of the disease highlights the urgent need for innovative therapeutic options. Immunotherapy can afford efficient and specific targeting of tumor cells, improving efficacy and reducing the side effects of current therapies. We have previously reported the aberrant expression of cancer/testis antigens (CTAs) in tumors of unrelated histological origin. In this study we investigated the expression and immunogenicity of the CTAs, Sperm Protein 17 (SP17), A-kinase anchor protein 4 (AKAP4) and Pituitary Tumor Transforming Gene 1 (PTTG1) in human non-small cell lung cancer (NSCLC) cell lines and primary tumors. We found that SP17, AKAP4 and PTTG1 are aberrantly expressed in cancer samples, compared to normal lung cell lines and tissues. We established the immunogenicity of these CTAs by measuring CTA-specific autoantibodies in patients' sera and generating CTA-specific autologous cytotoxic lymphocytes from patients' peripheral blood mononuclear cells. Our results provide proof of principle that the CTAs SP17/AKAP4/PTTG1 are expressed in both human NSCLC cell lines and primary tumors and can elicit an immunogenic response in lung cancer patients.
Collapse
MESH Headings
- A Kinase Anchor Proteins/genetics
- A Kinase Anchor Proteins/immunology
- A Kinase Anchor Proteins/metabolism
- Aged
- Aged, 80 and over
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Autoantibodies/blood
- Calmodulin-Binding Proteins
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carrier Proteins/genetics
- Carrier Proteins/immunology
- Carrier Proteins/metabolism
- Case-Control Studies
- Cell Line, Tumor
- Coculture Techniques
- Cytotoxicity, Immunologic
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunotherapy/methods
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Male
- Membrane Proteins
- Middle Aged
- RNA, Messenger/metabolism
- Securin/genetics
- Securin/immunology
- Securin/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Leonardo Mirandola
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA
| | - Jose A. Figueroa
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
- Kiromic, LLC. Lubbock, TX, USA
| | - Tam T. Phan
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
| | - Fabio Grizzi
- Humanitas Clinical and Research Center, Milano, Italy
| | - Minji Kim
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
| | | | - Marjorie R. Jenkins
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA
| | - Everardo Cobos
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA
- Kiromic, LLC. Lubbock, TX, USA
| | - Cynthia Jumper
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Raed Alalawi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Maurizio Chiriva-Internati
- Division of Hematology & Oncology and Southwest Cancer Treatment and Research Center, Texas Tech University, Lubbock, TX, USA
- Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA
- Kiromic, LLC. Lubbock, TX, USA
| |
Collapse
|
33
|
Ding M, Yang J. Therapeutic vaccination for non-small-cell lung cancer: a meta-analysis. Med Oncol 2014; 31:928. [PMID: 24647788 DOI: 10.1007/s12032-014-0928-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/12/2014] [Indexed: 10/25/2022]
Abstract
Lung cancer is the most common malignancy worldwide in terms of incidence and mortality. The vast majority of cases (85-90%) are non-small-cell lung cancer (NSCLC). Immunotherapy consists of mainly therapeutic vaccination designed to induce or amplify the immune responses directed against tumor-associated antigens. However, there is no conclusion to date for its strengths and weaknesses. Assessing the objective efficacy and safety of the therapeutic vaccination for NSCLC patients will help to figure out the future development of therapeutic vaccination. We performed a meta-analysis of six randomized controlled trials including 2,239 patients (1,363 patients in the therapeutic vaccination group and 876 patients in the control group) with NSCLC. Quantitative analysis was carried out to evaluate overall survival (OS) and toxicity of therapeutic vaccination. The vaccine group had produced significant improvement in OS compared with the control group [hazard ratio (HR) 0.83, 95% (confidence interval) CI 0.76-0.91; Z = 3.79, P = 0.0002]. Subgroup analysis showed a more significant improvement of OS in the subgroup compared with the control group (HR 0.70, 95% CI 0.59-0.82; Z = 4.42, P < 0.00001). No increased incidence of adverse events was obtained in the therapeutic vaccination group compared with the control group. Therapeutic vaccination adds benefits to NSCLC patients and may become a standard complementary therapeutic approach in the future if the associated toxicity is reduced.
Collapse
Affiliation(s)
- Mei Ding
- Zhongnan Hospital of Wuhan University, Wuhan, China,
| | | |
Collapse
|
34
|
Tumor-induced CD8+ T-cell dysfunction in lung cancer patients. Clin Dev Immunol 2012; 2012:741741. [PMID: 23118782 PMCID: PMC3483679 DOI: 10.1155/2012/741741] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/28/2012] [Accepted: 09/04/2012] [Indexed: 12/21/2022]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide and one of the most common types of cancers. The limited success of chemotherapy and radiotherapy regimes have highlighted the need to develop new therapies like antitumor immunotherapy. CD8+ T-cells represent a major arm of the cell-mediated anti-tumor response and a promising target for developing T-cell-based immunotherapies against lung cancer. Lung tumors, however, have been considered to possess poor immunogenicity; even so, lung tumor-specific CD8+ T-cell clones can be established that possess cytotoxicity against autologous tumor cells. This paper will focus on the alterations induced in CD8+ T-cells by lung cancer. Although memory CD8+ T-cells infiltrate lung tumors, in both tumor-infiltrating lymphocytes (TILs) and malignant pleural effusions, these cells are dysfunctional and the effector subset is reduced. We propose that chronic presence of lung tumors induces dysfunctions in CD8+ T-cells and sensitizes them to activation-induced cell death, which may be associated with the poor clinical responses observed in immunotherapeutic trials. Getting a deeper knowledge of the evasion mechanisms lung cancer induce in CD8+ T-cells should lead to further understanding of lung cancer biology, overcome tumor evasion mechanisms, and design improved immunotherapeutic treatments for lung cancer.
Collapse
|
35
|
Abstract
To date, in lung cancer, early attempts to modulate the immune system via vaccine-based therapeutics have been unsuccessful. An improved understanding of tumor immunology has facilitated the production of more sophisticated lung cancer vaccines. It is anticipated that it will likely require multiple epitopes of a diverse set of genes restricted to multiple haplotypes to generate a truly effective vaccine that is able to overcome the various immunologic escape mechanisms that tumors employ. Other issues to overcome include optimal patient selection, which adjuvant agent to use, and how to adequately monitor for an immunologic response. This review discusses the most promising vaccination strategies for non-small cell lung cancer including the allogeneic tumor cell vaccine belagenpumatucel-L, which is a mixture of 4 allogeneic non-small cell lung cancer cell lines genetically modified to secrete an antisense oligonucleotide to transforming growth factor β2 and 3 other target protein-specific vaccines designed to induce responses against melanoma-associated antigen A3, mucin 1, and epidermal growth factor.
Collapse
|
36
|
Wang J, Zou ZH, Xia HL, He JX, Zhong NS, Tao AL. Strengths and weaknesses of immunotherapy for advanced non-small-cell lung cancer: a meta-analysis of 12 randomized controlled trials. PLoS One 2012; 7:e32695. [PMID: 22403699 PMCID: PMC3293858 DOI: 10.1371/journal.pone.0032695] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 01/30/2012] [Indexed: 12/31/2022] Open
Abstract
Background Lung cancer is one of the leading causes of cancer death worldwide. Non-small-cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancers. Immunotherapy has yielded no consistent benefit to date for those patients. Assessing the objective efficacy and safety of immunotherapy for advanced NSCLC patients will help to instruct the future development of immunotherapeutic drugs. Methodology and Principal Findings We performed a meta-analysis of 12 randomized controlled trials including 3134 patients (1570 patients in the immunotherapy group and 1564 patients in the control group) with histologically confirmed stage IIIA, IIIB, or IV NSCLC. The analysis was executed with efficacy end points regarding overall survival (OS), progression-free survival (PFS), complete response (CR), partial response (PR), and total effective rate. Overall unstratified OS, PFS, PR, and total effective rate were significantly improved in advanced NSCLC patients in the immunotherapy group (P = 0.0007, 0.0004, 0.002, 0.003, respectively), whereas CR was not improved (P = 0.97). Subgroup analysis showed that monoclonal antibody (mAb) immunotherapy significantly improved the PFS, PR, and total effective rate and showed a trend of improving OS of advanced NSCLC patients compared with the control group, with one kind of adverse event being significantly dominant. Compared with the control group, the vaccine subgroup showed no significant difference with regard to serious adverse events, whereas cytokine immunotherapy significantly induced three kinds of serious adverse events. Conclusions Immunotherapy works efficiently on advanced NSCLC patients. Of several immunotherapies, mAb therapy may be a potential immunotherapy for advanced NSCLC patients, and become a standard complementary therapeutic approach in the future if the issues concerning toxicity and allergenicity of mAbs have been overcome.
Collapse
Affiliation(s)
- Juan Wang
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ze-Hong Zou
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong-Lin Xia
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian-Xing He
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nan-Shan Zhong
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ai-Lin Tao
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- * E-mail:
| |
Collapse
|
37
|
Tucker ZCG, Laguna BA, Moon E, Singhal S. Adjuvant immunotherapy for non-small cell lung cancer. Cancer Treat Rev 2012; 38:650-61. [PMID: 22226940 DOI: 10.1016/j.ctrv.2011.11.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 11/28/2011] [Accepted: 11/30/2011] [Indexed: 01/20/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the biggest cancer killer in the United States and worldwide. In 2011, there are estimated to be 221,130 new cases of lung cancer in the United States. Over a million people will die of lung cancer worldwide this year alone. When possible, surgery to remove the tumor is the best treatment strategy for patients with NSCLC. However, even with adjuvant (postoperative) chemotherapy and radiation, more than 40% of patients will develop recurrences locally or systemically and ultimately succumb to their disease. Thus, there is an urgent need for developing superior approaches to treat patients who undergo surgery for NSCLC to eliminate residual disease that is likely responsible for these recurrences. Our group and others have been interested in using immunotherapy to augment the efficacy of current treatment strategies. Immunotherapy is very effective against minimal disease burden and small deposits of tumor cells that are accessible by the circulating immune cells. Therefore, this strategy may be ideally suited as an adjunct to surgery to seek and destroy microscopic tumor deposits that remain after surgery. This review describes the mechanistic underpinnings of immunotherapy and how it is currently being used to target residual disease and prevent postoperative recurrences after pulmonary resection in NSCLC.
Collapse
Affiliation(s)
- Zachary C G Tucker
- Division of Thoracic Surgery, University of Pennsylvania School of Medicine, 6 White, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
38
|
Chen L, He Z, Qin L, Li Q, Shi X, Zhao S, Chen L, Zhong N, Chen X. Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity. PLoS One 2011; 6:e24407. [PMID: 21931708 PMCID: PMC3170332 DOI: 10.1371/journal.pone.0024407] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 08/08/2011] [Indexed: 12/22/2022] Open
Abstract
Background Lung cancer is the most common malignancy in humans and its high fatality means that no effective treatment is available. Developing new therapeutic strategies for lung cancer is urgently needed. Malaria has been reported to stimulate host immune responses, which are believed to be efficacious for combating some clinical cancers. This study is aimed to provide evidence that malaria parasite infection is therapeutic for lung cancer. Methodology/Principal Findings Antitumor effect of malaria infection was examined in both subcutaneously and intravenously implanted murine Lewis lung cancer (LLC) model. The results showed that malaria infection inhibited LLC growth and metastasis and prolonged the survival of tumor-bearing mice. Histological analysis of tumors from mice infected with malaria revealed that angiogenesis was inhibited, which correlated with increased terminal deoxynucleotidyl transferase-mediated (TUNEL) staining and decreased Ki-67 expression in tumors. Through natural killer (NK) cell cytotoxicity activity, cytokine assays, enzyme-linked immunospot assay, lymphocyte proliferation, and flow cytometry, we demonstrated that malaria infection provided anti-tumor effects by inducing both a potent anti-tumor innate immune response, including the secretion of IFN-γ and TNF-α and the activation of NK cells as well as adaptive anti-tumor immunity with increasing tumor-specific T-cell proliferation and cytolytic activity of CD8+ T cells. Notably, tumor-bearing mice infected with the parasite developed long-lasting and effective tumor-specific immunity. Consequently, we found that malaria parasite infection could enhance the immune response of lung cancer DNA vaccine pcDNA3.1-hMUC1 and the combination produced a synergistic antitumor effect. Conclusions/Significance Malaria infection significantly suppresses LLC growth via induction of innate and adaptive antitumor responses in a mouse model. These data suggest that the malaria parasite may provide a novel strategy or therapeutic vaccine vector for anti-lung cancer immune-based therapy.
Collapse
MESH Headings
- Adaptive Immunity/immunology
- Animals
- Apoptosis
- Cancer Vaccines/immunology
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/parasitology
- Carcinoma, Lewis Lung/surgery
- Cell Proliferation
- Cytokines/biosynthesis
- Cytotoxicity, Immunologic/immunology
- Dendritic Cells/immunology
- Disease Models, Animal
- Immunity, Innate/immunology
- Killer Cells, Natural/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Malaria/parasitology
- Mice
- Neoplasm Metastasis
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Parasites/immunology
- Plasmodium yoelii/immunology
- Th1 Cells/immunology
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Lili Chen
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS-FS Biotech and Pharmaceutical Center, Chinese Academy of Sciences, Foshan, China
| | - Zhengxiang He
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS-FS Biotech and Pharmaceutical Center, Chinese Academy of Sciences, Foshan, China
| | - Li Qin
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS-FS Biotech and Pharmaceutical Center, Chinese Academy of Sciences, Foshan, China
| | - Qinyan Li
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS-FS Biotech and Pharmaceutical Center, Chinese Academy of Sciences, Foshan, China
| | - Xibao Shi
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Siting Zhao
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ling Chen
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- * E-mail: (XC) (NZ); (NZ) (XC)
| | - Xiaoping Chen
- Center for Infection and Immunity, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS-FS Biotech and Pharmaceutical Center, Chinese Academy of Sciences, Foshan, China
- * E-mail: (XC) (NZ); (NZ) (XC)
| |
Collapse
|
39
|
Mellstedt H, Vansteenkiste J, Thatcher N. Vaccines for the treatment of non-small cell lung cancer: Investigational approaches and clinical experience. Lung Cancer 2011; 73:11-7. [DOI: 10.1016/j.lungcan.2011.02.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 02/04/2011] [Accepted: 02/06/2011] [Indexed: 11/17/2022]
|
40
|
Immunotherapy for lung cancers. J Biomed Biotechnol 2011; 2011:250860. [PMID: 21318107 PMCID: PMC3035001 DOI: 10.1155/2011/250860] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/15/2010] [Accepted: 12/23/2010] [Indexed: 11/21/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although treatment methods in surgery, irradiation, and chemotherapy have improved, prognosis remains unsatisfactory and developing new therapeutic strategies is still an urgent demand. Immunotherapy is a novel therapeutic approach wherein activated immune cells can specifically kill tumor cells by recognition of tumor-associated antigens without damage to normal cells. Several lung cancer vaccines have demonstrated prolonged survival time in phase II and phase III trials, and several clinical trials are under investigation. However, many clinical trials involving cancer vaccination with defined tumor antigens work in only a small number of patients. Cancer immunotherapy is not completely effective in eradicating tumor cells because tumor cells escape from host immune scrutiny. Understanding of the mechanism of immune evasion regulated by tumor cells is required for the development of more effective immunotherapeutic approaches against lung cancer. This paper discusses the identification of tumor antigens in lung cancer, tumor immune escape mechanisms, and clinical vaccine trials in lung cancer.
Collapse
|
41
|
Levitzki A, Klein S. Signal transduction therapy of cancer. Mol Aspects Med 2010; 31:287-329. [DOI: 10.1016/j.mam.2010.04.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Accepted: 04/28/2010] [Indexed: 01/05/2023]
|
42
|
Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol 2010; 28:682-8. [PMID: 20373055 DOI: 10.1007/s12032-010-9515-2] [Citation(s) in RCA: 483] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 03/22/2010] [Indexed: 01/06/2023]
Abstract
The immunohistochemical analysis was used to evaluate the expression of PD-L1 in 109 non-small cell lung cancer (NSCLC) tissues and para-tumor tissues. Associations between expressed PD-L1 and tumor histological types, degree of differentiation, and lymph node metastasis were calculated, and overall survival was assessed. Meanwhile, immunohistochemistry and immunofluorescence double labeling technique were performed to detect the expressions of PD-L1, CD1α, and CD83 on TIDC of 20 lung cancer tissues, and the expression of PD-L1 in CD1α+DCs and CD83+DCs and their significances were also explored. We found that the expression rate of PD-L1 in NSCLC was associated with histological types and overall survival. Patients with either adenocarcinoma or survival time after surgery less than 3 years showed higher expression rate of PD-L1. Furthermore, Cox model analysis indicated that PD-L1 might be regarded as a poor prognostic factor. PD-L1 could be also detected in CD1α+ immature DC in NSCLC, indicating that as a class of key anti-tumor immunocyte in tumor microenvironment, DC expressing PD-L1 itself might play an important role in keeping its immature status and contributing to tumor cells immune escape and disease progression.
Collapse
Affiliation(s)
- Chuan-Yong Mu
- Respiratory Department, The First Affiliated Hospital of Soochow University, 188 Shizi Street, 215006, Suzhou, China
| | | | | | | | | |
Collapse
|