101
|
Su C, Wang H, Liu Y, Guo Q, Zhang L, Li J, Zhou W, Yan Y, Zhou X, Zhang J. Adverse Effects of Anti-PD-1/PD-L1 Therapy in Non-small Cell Lung Cancer. Front Oncol 2020; 10:554313. [PMID: 33072580 PMCID: PMC7531287 DOI: 10.3389/fonc.2020.554313] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022] Open
Abstract
Currently, immunotherapy has shown great efficacy in clinical trials, and monoclonal antibodies directed against immune checkpoint PD-1/PD-L1 have shown encouraging results in first-line or second-line treatment of non-small cell lung cancer patients. Meanwhile, anti-PD-1/PD-L1 immune checkpoint drugs combined with other treatments, such as chemotherapy, targeted therapy as well as anti-CTLA-4 checkpoint therapy, are considered an attractive treatment with higher efficacy. However, toxicity associated with PD-1/PD-L1 blockade is worth attention. Understanding the adverse effects caused by anti-PD-1/PD-L1 immunosuppressive agents is vital to guide the clinical rational use of drug. In this review, we summarized the adverse effects that occurred during the clinical use of anti-PD-1/PD-L1 inhibitors in the treatment of non-small cell lung cancer and discussed how to effectively manage and respond to these adverse reactions.
Collapse
Affiliation(s)
- Chaoyue Su
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,School of Public Health, Hainan Medical University, Haikou, China
| | - Hui Wang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yunru Liu
- School of Public Health, Hainan Medical University, Haikou, China
| | - Qiaoru Guo
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lingling Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jiajun Li
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenmin Zhou
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yanyan Yan
- Institute of Immunology and School of Medicine, Shanxi Datong University, Datong, China
| | - Xinke Zhou
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jianye Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,School of Public Health, Hainan Medical University, Haikou, China
| |
Collapse
|
102
|
Singh N, Seetharamu N. Complete response with anti-PD-L1 antibody following progression on anti-PD-1 antibody in advanced non-small cell lung cancer. BMJ Case Rep 2020; 13:13/8/e236101. [PMID: 32843385 DOI: 10.1136/bcr-2020-236101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Immune-checkpoint inhibitors (ICI), specifically inhibitors of programmed death ligand-1 (PD-L1) and receptor (PD-1) are the new standard of care for the treatment of patients with advanced non-small cell lung cancer (NSCLC) in front line setting as monotherapy or along with chemotherapy. Many of these agents are also approved for use in subsequent lines of treatment on progression on platinum doublet chemotherapy. Nivolumab, pembrolizumab and atezolizumab are currently approved ICI for advanced NSCLC. To date, no study has reported efficacy and safety of alternate PD-1/PD-L1 inhibitors in patients with NSCLC who have progressed on one ICI. Here, we report a case of a patient with advanced NSCLC who had a complete response to atezolizumab, following progression of disease on platinum doublet chemotherapy and then, nivolumab monotherapy.
Collapse
Affiliation(s)
- Navdeep Singh
- Department of Medicine, North Shore University Hospital at Manhasset, Manhasset, New York, USA
| | - Nagashree Seetharamu
- Department of Medicine/Division of Hematology and Medical Oncology, Northwell Health, Lake Success, New York, USA.,Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
103
|
Li C, Liu H, Niu Q, Gao J. Circ_0000376, a Novel circRNA, Promotes the Progression of Non-Small Cell Lung Cancer Through Regulating the miR-1182/NOVA2 Network. Cancer Manag Res 2020; 12:7635-7647. [PMID: 32922073 PMCID: PMC7455537 DOI: 10.2147/cmar.s258340] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Background Hypoxia has been shown to induce the malignant progression of cancer, including non-small cell lung cancer (NSCLC). Circular RNA (circRNA) is considered to be an important regulator of cancer progression. However, the role of a newly discovered circRNA, circ_0000376, in the progression of NSCLC is unclear. Methods The relative expression levels of circ_0000376, miR-1182 and neuro-oncological ventral antigen 2 (NOVA2) were detected via quantitative real-time polymerase chain reaction (qRT-PCR). Glucose consumption and lactate production were determined using Glucose Assay Kit and Lactate Assay Kit, respectively. Moreover, the protein levels of glycolysis markers and NOVA2 were measured using Western blot (WB) analysis. Furthermore, 3-(4, 5-dimethyl-2 thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay was performed to assess cell viability, and transwell assay was employed to evaluate cell migration and invasion. The interaction between miR-1182 and circ_0000376 or NOVA2 was confirmed by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. In addition, animal experiments were conducted to assess the influence of circ_0000376 silencing on NSCLC tumor growth in vivo. Results Circ_0000376 was upregulated in NSCLC, and its high expression was related to the poor overall survival of NSCLC patients. Hypoxia could enhance circ_0000376 expression and promote the glycolysis, viability, migration, and invasion of NSCLC cells. However, silencing of circ_0000376 could inhibit the glycolysis, viability, migration, and invasion of hypoxia-induced NSCLC cells. Additionally, circ_0000376 could sponge miR-1182, and miR-1182 could target NOVA2. MiR-1182 silencing could reverse the inhibitory effect of circ_0000376 knockdown on NSCLC progression, and NOVA2 overexpression also could reverse the suppressive effect of miR-1182 overexpression on NSCLC progression. Meanwhile, miR-1182 inhibitor could invert the negative regulation effect of circ_0000376 silencing on NOVA2 expression. In addition, circ_0000376 knockdown inhibited the NSCLC tumor growth via regulating the miR-1182 and NOVA2 expression in vivo. Conclusion Circ_0000376 promoted NSCLC progression by regulating the miR-1182/NOVA2 axis, suggesting that circ_0000376 might be a potential biomarker for NSCLC treatment.
Collapse
Affiliation(s)
- Cui Li
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Hai Liu
- Department of Clinical Laboratory, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Qin Niu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Jia Gao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| |
Collapse
|
104
|
Kim JY, Choi JK, Jung H. Genome-wide methylation patterns predict clinical benefit of immunotherapy in lung cancer. Clin Epigenetics 2020; 12:119. [PMID: 32762727 PMCID: PMC7410160 DOI: 10.1186/s13148-020-00907-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background It is crucial to unravel molecular determinants of responses to immune checkpoint blockade (ICB) therapy because only a small subset of advanced non-small cell lung cancer (NSCLC) patients responds to ICB therapy. Previous studies were concentrated on genomic and transcriptomic markers (e.g., mutation burden and immune gene expression). However, these markers are not sufficient to accurately predict a response to ICB therapy. Results Here, we analyzed DNA methylomes of 141 advanced NSCLC samples subjected to ICB therapy (i.e., anti-programmed death-1) from two independent cohorts (60 and 81 patients from our and IDIBELL cohorts). Integrative analysis of patients with matched transcriptome data in our cohort (n = 28) at pathway level revealed significant overlaps between promoter hypermethylation and transcriptional repression in nonresponders relative to responders. Fifteen immune-related pathways, including interferon signaling, were identified to be enriched for both hypermethylation and repression. We built a reliable prognostic risk model based on eight genes using LASSO model and successfully validated the model in independent cohorts. Furthermore, we found 30 survival-associated molecular interaction networks, in which two or three hypermethylated genes showed significant mutual exclusion across nonresponders. Conclusions Our study demonstrates that methylation patterns can provide insight into molecular determinants underlying the clinical benefit of ICB therapy.
Collapse
Affiliation(s)
- Jeong Yeon Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea. .,Penta Medix Co., Ltd., Seongnam-si, Gyeongi-do, 13449, Republic of Korea.
| | - Hyunchul Jung
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea. .,Cancer Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Cambridge, UK.
| |
Collapse
|
105
|
James NE, Woodman M, DiSilvestro PA, Ribeiro JR. The Perfect Combination: Enhancing Patient Response to PD-1-Based Therapies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E2150. [PMID: 32756436 PMCID: PMC7466102 DOI: 10.3390/cancers12082150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, with an overall 5-year survival of only 47%. As the development of novel targeted therapies is drastically necessary in order to improve patient survival, current EOC clinical trials have heavily focused on immunotherapeutic approaches, centered upon programmed cell death 1 (PD-1) inhibitors. While PD-1 monotherapies have only exhibited modest responses for patients, it has been theorized that in order to enhance EOC patient response to immunotherapy, combinatorial regimens must be investigated. In this review, unique challenges to EOC PD-1 response will be discussed, along with a comprehensive description of both preclinical and clinical studies evaluating PD-1-based combinatorial therapies. Promising aspects of PD-1-based combinatorial approaches are highlighted, while also discussing specific preclinical and clinical areas of research that need to be addressed, in order to optimize EOC patient immunotherapy response.
Collapse
Affiliation(s)
- Nicole E. James
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Morgan Woodman
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Paul A. DiSilvestro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| | - Jennifer R. Ribeiro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| |
Collapse
|
106
|
Wu Z, Man S, Sun R, Li Z, Wu Y, Zuo D. Recent advances and challenges of immune checkpoint inhibitors in immunotherapy of non-small cell lung cancer. Int Immunopharmacol 2020; 85:106613. [DOI: 10.1016/j.intimp.2020.106613] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
|
107
|
Zhong S, Cui Y, Liu Q, Chen S. CAR-T cell therapy for lung cancer: a promising but challenging future. J Thorac Dis 2020; 12:4516-4521. [PMID: 32944366 PMCID: PMC7475572 DOI: 10.21037/jtd.2020.03.118] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/20/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Shupeng Zhong
- Department of Oncology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Treatment, Guangzhou 510080, China
| | - Yingqiang Cui
- Department of Oncology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Treatment, Guangzhou 510080, China
| | - Qian Liu
- Central Laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Treatment, Guangzhou 510080, China
| | - Size Chen
- Department of Oncology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Treatment, Guangzhou 510080, China
| |
Collapse
|
108
|
Dhillon S, Syed YY. Atezolizumab First-Line Combination Therapy: A Review in Metastatic Nonsquamous NSCLC. Target Oncol 2020; 14:759-768. [PMID: 31728860 DOI: 10.1007/s11523-019-00686-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atezolizumab (Tecentriq®), a humanized, anti-programmed cell death ligand-1 (PD-L1) monoclonal antibody, in combination with bevacizumab, carboplatin and paclitaxel (ABCP) or with carboplatin and nab-paclitaxel (ACnP) has been approved as first-line treatment for metastatic nonsquamous NSCLC, based on results from the randomized IMpower150 and IMpower130 studies in chemotherapy-naïve patients with nonsquamous metastatic NSCLC. In IMpower150, ABCP prolonged progression-free survival (PFS) and overall survival (OS) relative to BCP, regardless of EGFR or ALK status, liver metastases at baseline or PD-L1 expression levels. In IMpower130, ACnP prolonged PFS and OS relative to CnP in patients without EGFR or ALK genetic aberrations. ABCP and ACnP had manageable tolerability profiles, which were consistent with the profile of the individual components of the regimen. Immune-related adverse events with ABCP and ACnP were largely mild or moderate in severity, and most were reversible with interruption of atezolizumab and initiation of appropriate treatment. Current evidence indicates that ABCP and ACnP are valuable emerging first-line treatment options for metastatic nonsquamous NSCLC.
Collapse
Affiliation(s)
- Sohita Dhillon
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand
| | - Yahiya Y Syed
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
109
|
Chen Y, Pei Y, Luo J, Huang Z, Yu J, Meng X. Looking for the Optimal PD-1/PD-L1 Inhibitor in Cancer Treatment: A Comparison in Basic Structure, Function, and Clinical Practice. Front Immunol 2020; 11:1088. [PMID: 32547566 PMCID: PMC7274131 DOI: 10.3389/fimmu.2020.01088] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/05/2020] [Indexed: 12/18/2022] Open
Abstract
Programmed cell death protein-1/ligand 1 (PD-1/L1) targeted immune checkpoint inhibitors have become the focus of tumor treatment due to their promising efficacy. Currently, several PD-1/PD-L1 inhibitors have been approved for clinical practice with several more in clinical trials. Notably, based on available trial data, the selection of different PD-1/PD-L1 inhibitors in the therapeutic application and the corresponding efficacy varies. Widespread attention then is increasingly raised to the clinical comparability of different PD-1/PD-L1 inhibitors. The comparison of the inhibitors could not only help clinicians make in-depth understanding of them, but also further facilitate the selection of the optimal inhibitor for patients in treatment as well as for future clinical research and the development of new related drugs. As we all know, molecular structure could determine molecular function, which further affects their application. Therefore, in this review, we aim to comprehensively compare the structural basis, molecular biological functions, and clinical practice of different PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yu Chen
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yanqing Pei
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jingyu Luo
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhaoqin Huang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jinming Yu
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiangjiao Meng
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
110
|
Hu J, Wang Z, Chen Z, Li A, Sun J, Zheng M, Wu J, Shen T, Qiao J, Li L, Li B, Wu D, Xiao Q. DKK2 blockage-mediated immunotherapy enhances anti-angiogenic therapy of Kras mutated colorectal cancer. Biomed Pharmacother 2020; 127:110229. [PMID: 32559853 PMCID: PMC7523634 DOI: 10.1016/j.biopha.2020.110229] [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] [Received: 03/30/2020] [Revised: 04/26/2020] [Accepted: 05/03/2020] [Indexed: 02/07/2023] Open
Abstract
There are limited options for targeted therapies for colorectal cancer (CRC). Anti-EGFR therapy is limited to CRC without KRAS mutations. Even worse, most of CRC are refractory to currently immune checkpoint blockade. DKK2, which is upregulated in CRC, was recently found to suppress host immune responses, and its blockage effectively impeded tumor progression in benign genetic CRC models in our previous study. Here, our recent study demonstrated that in human CRC tumor samples expressing high levels of DKK2, DKK2 blockade caused stronger activation of tumor infiltrating CD8+ T cells in ex vivo culture. Intriguingly, we observed a correlation of high DKK2 expression with increased lymph node metastasis prevalence in these CRC patients as well. Furthermore, in a mouse genetic CRC model with mutations in APC and KRAS, which more closely mimics advanced human CRC, we confirmed the tumor inhibitory effect of DKK2 blockade, which significantly retarded tumor progression and extended survival, with increased immune effector cell activation and reduced angiogenesis. Based on this, we performed a combined administration of DKK2 blockade with sub-optimal anti-VEGFR treatment and observed a synergetic effect on suppressing tumor angiogenesis and progression, as well as extending survival, better than those of every single therapy. Thus, this study provides further evidence for the potential therapeutic application of DKK2 blockade in the clinical treatment of human CRC.
Collapse
Affiliation(s)
- Jiajia Hu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengxi Chen
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States; Department of Orthodontics, Shanghai Ninth People׳s Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Ao Li
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jibo Wu
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Tianli Shen
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States; Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Ju Qiao
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, United States
| | - Lin Li
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dianqing Wu
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States.
| | - Qian Xiao
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
111
|
Liang ZZ, Guo C, Zou MM, Meng P, Zhang TT. circRNA-miRNA-mRNA regulatory network in human lung cancer: an update. Cancer Cell Int 2020; 20:173. [PMID: 32467668 PMCID: PMC7236303 DOI: 10.1186/s12935-020-01245-4] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Circular RNAs, as hopeful diagnosis markers and therapeutic molecules, have been studied, probed and applied into several diseases, such as cardiovascular diseases, systemic lupus erythematosus, leukemia, pulmonary tuberculosis, and cancer especially. Recently, mounting evidence has supported that circRNAs play a key role in the tumorigenesis, progress, invasion and metastasis in lung cancer. Its special structure—3′–5′ covalent loop—allow it to execute several special functions in both normal eukaryotic cells and cancer cells. Our review summaries the latest studies on characteristics and biogenesis of circRNAs, and highlight the regulatory functions about miRNA sponge of lung-cancer-related circRNAs. In addition, the interaction of the circRNA-miRNA-mRNA regulatory network will also be elaborated in detail in this review. Therefore, this review can provide a new idea or strategy for further development and application in clinical setting in terms of early-diagnosis and better treatment.
Collapse
Affiliation(s)
- Zhuo-Zheng Liang
- 1Department of Pulmonary Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Cheng Guo
- 2Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Man-Man Zou
- 1Department of Pulmonary Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Ping Meng
- 1Department of Pulmonary Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Tian-Tuo Zhang
- 1Department of Pulmonary Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| |
Collapse
|
112
|
Rossi G, Russo A, Tagliamento M, Tuzi A, Nigro O, Vallome G, Sini C, Grassi M, Dal Bello MG, Coco S, Longo L, Zullo L, Tanda ET, Dellepiane C, Pronzato P, Genova C. Precision Medicine for NSCLC in the Era of Immunotherapy: New Biomarkers to Select the Most Suitable Treatment or the Most Suitable Patient. Cancers (Basel) 2020; 12:E1125. [PMID: 32365882 PMCID: PMC7281184 DOI: 10.3390/cancers12051125] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, the evolution of treatments has made it possible to significantly improve the outcomes of patients with non-small cell lung cancer (NSCLC). In particular, while molecular targeted therapies are effective in specific patient sub-groups, immune checkpoint inhibitors (ICIs) have greatly influenced the outcomes of a large proportion of NSCLC patients. While nivolumab activity was initially assessed irrespective of predictive biomarkers, subsequent pivotal studies involving other PD-1/PD-L1 inhibitors in pre-treated advanced NSCLC (atezolizumab within the OAK study and pembrolizumab in the Keynote 010 study) reported the first correlations between clinical outcomes and PD-L1 expression. However, PD-L1 could not be sufficient on its own to select patients who may benefit from immunotherapy. Many studies have tried to discover more precise markers that are derived from tumor tissue or from peripheral blood. This review aims to analyze any characteristics of the immunogram that could be used as a predictive biomarker for response to ICIs. Furthermore, we describe the most important genetic alteration that might predict the activity of immunotherapy.
Collapse
Affiliation(s)
- Giovanni Rossi
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | | | - Marco Tagliamento
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Alessandro Tuzi
- UO Oncologia, ASST Sette Laghi, 21100 Varese, Italy; (A.T.); (O.N.)
| | - Olga Nigro
- UO Oncologia, ASST Sette Laghi, 21100 Varese, Italy; (A.T.); (O.N.)
| | - Giacomo Vallome
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Claudio Sini
- Oncologia Medica e CPDO, ASSL di Olbia-ATS Sardegna, 07026 Olbia, Italy;
| | - Massimiliano Grassi
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Simona Coco
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Luca Longo
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Lodovica Zullo
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Enrica Teresa Tanda
- Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Chiara Dellepiane
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Paolo Pronzato
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Carlo Genova
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| |
Collapse
|
113
|
Challenges to Successful Implementation of the Immune Checkpoint Inhibitors for Treatment of Glioblastoma. Int J Mol Sci 2020; 21:ijms21082759. [PMID: 32316096 PMCID: PMC7215941 DOI: 10.3390/ijms21082759] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant glioma, treatment of which has not improved significantly in many years. This is due to the unique challenges that GBM tumors present when designing and implementing therapies. Recently, immunotherapy in the form of immune checkpoint inhibition (ICI) has revolutionized the treatment of various malignancies. The application of immune checkpoint inhibition in GBM treatment has shown promising preclinical results. Unfortunately, this has met with little to no success in the clinic thus far. In this review, we will discuss the challenges presented by GBM tumors that likely limit the effect of ICI and discuss the approaches being tested to overcome these challenges.
Collapse
|
114
|
Käsmann L, Janssen S, Baschnagel AM, Kruser TJ, Harada H, Aktan M, Rades D. Prognostic factors and outcome of reirradiation for locally recurrent small cell lung cancer-a multicenter study. Transl Lung Cancer Res 2020; 9:232-238. [PMID: 32420062 PMCID: PMC7225148 DOI: 10.21037/tlcr.2020.01.19] [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: 12/23/2022]
Abstract
Background The prognosis of patients with recurrent small cell lung cancer (SCLC) remains poor and treatment options are limited. We performed a multi-institution retrospective cohort study to evaluate the outcome of thoracic reirradiation, identify prognostic factors and assess treatment-related toxicity. Methods Data of 33 patients re-irradiated for recurrent SCLC at 4 international university hospitals, were analysed. Overall survival (OS) acute and late toxicities were evaluated and prognostic factors for reirradiation were identified. Results Reirradiation (Re-RT) was performed at a median interval time of 24 months after the first thoracic radiotherapy series. Median survival after reirradiation was 7 months (range, 1-54 months). The Re-RT dose in EQD2 ranged from 20 to 87.50 Gy with a median of 32.50 Gy. The 1- and 2-year OS were 33% and 17%, respectively. Patients with a good performance status (KPS >70%), absence of extrathoracic disease, reirradiation dose (EQD2) of >40 Gy and a cumulative dose of first plus second series of radiotherapy (EQD2) >90 Gy were associated with improved OS. Acute pulmonary Grade 1-2 toxicity from re-irradiation was recorded in 11 patients (33%) and grade 3 acute toxicity was encountered 1 patient (3%). Conclusions Reirradiation for locoregionally recurrent SCLC is safe and shows promising outcomes. Patients reirradiated with doses >40 Gy experienced more favourable survival rates. In contrast, patients with a poor performance status or extrathoracic disease have a poor prognosis and Re-RT should be considered only for symptom control in this group.
Collapse
Affiliation(s)
- Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany.,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Stefan Janssen
- Medical Practice for Radiotherapy and Radiation Oncology, Hannover, Germany.,Department of Radiation Oncology, University of Lübeck, Lübeck, Germany
| | - Andrew M Baschnagel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Tim J Kruser
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hideyuki Harada
- Division of Radiation Therapy, Shizuoka Cancer Center, 1007 Shimonagakubo, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Meryem Aktan
- Department of Radiation Oncology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Dirk Rades
- Department of Radiation Oncology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
115
|
He J, Pan Y, Guo Y, Li B, Tang Y. Study on the Expression Levels and Clinical Significance of PD-1 and PD-L1 in Plasma of NSCLC Patients. J Immunother 2020; 43:156-164. [DOI: 10.1097/cji.0000000000000315] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
116
|
Qin T, Xia J, Liu S, Wang J, Liu H, Zhang Y, Jia Y, Li K. Clinical importance of VEGFC and PD-L1 co-expression in lung adenocarcinoma patients. Thorac Cancer 2020; 11:1139-1148. [PMID: 32154654 PMCID: PMC7180596 DOI: 10.1111/1759-7714.13354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 12/25/2022] Open
Abstract
Background Vascular endothelial growth factor C (VEGFC), an activator of lymphangiogenesis, is newly identified as an immunomodulator which can regulate the immune system so that tumor cells more easily escape immune surveillance. Evidence has shown programmed cell death‐ligand 1 (PD‐L1) can also suppress the immune response. Nevertheless, the clinical significance of co‐expression of VEGFC and PD‐L1 for predicting outcomes in patients with lung adenocarcinoma has not yet been determined. Methods A total of 114 patients with lung adenocarcinoma who underwent surgeries at Tianjin Medical University Cancer Institute and Hospital between December 2011 and September 2016 were retrospectively reviewed. Tissue specimens were collected for immunohistochemistry of VEGFC and PD‐L1 which were analyzed with an H‐score system. Results In this study, 57 (50.0%) and 47 (41.2%) patients were classified as VEGFC high expression and PD‐L1 high expression. Co‐expression was observed in 33 (28.9%) patients. In addition, a positive correlation was found between VEGFC and PD‐L1 (P = 0.0398, r = 0.1937). In a univariate analysis, both progression‐free survival (PFS) and overall survival (OS) were significantly worse in the VEGFC high expression group and the PD‐L1 high expression group, respectively. Furthermore, VEGFC/PD‐L1 co‐expression showed a worse OS (P = 0.03) and PFS survival (P = 0.01) than the other groups. Conclusions Taken together, these results indicate that VEGFC/PD‐L1 co‐expression can forecast both poor OS and PFS in patients with resected lung adenocarcinoma. Co‐expression of VEGFC and PD‐L1 may serve as a significant prognostic factor for patients with lung adenocarcinoma. Key points VEGFC/PD‐L1 co‐expression forecasts poor survival in patients with resected lung adenocarcinoma. VEGFC/PD‐L1 co‐expression may be used as a prognostic indicator and provide the theoretical possibility to screen the optimal population with a combination of anti‐VEGFC and anti‐PD‐L1 therapy in the clinical treatment.
Collapse
Affiliation(s)
- Tingting Qin
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| | - Junling Xia
- Department of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Shaochuan Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| | - Jing Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| | - Hailin Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| | - Yan Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| | - Yanan Jia
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| | - Kai Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
117
|
Gennen K, Käsmann L, Taugner J, Eze C, Karin M, Roengvoraphoj O, Neumann J, Tufman A, Orth M, Reu S, Belka C, Manapov F. Prognostic value of PD-L1 expression on tumor cells combined with CD8+ TIL density in patients with locally advanced non-small cell lung cancer treated with concurrent chemoradiotherapy. Radiat Oncol 2020; 15:5. [PMID: 31898519 PMCID: PMC6941268 DOI: 10.1186/s13014-019-1453-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022] Open
Abstract
Background/aim mmune checkpoint inhibition (CPI) has an increasing impact in the multimodal treatment of locally advanced non-small cell lung cancer (LA-NSCLC). Increasing evidence suggests treatment outcome depending on tumor cell PD-L1 expression. The purpose of this retrospective study was to investigate the prognostic value of PD-L1 expression on tumor cells in combination with CD8+ tumor stroma-infiltrating lymphocyte (TIL) density in inoperable LA-NSCLC treated with concurrent chemoradiotherapy (CRT). Patients and method We retrospectively assessed clinical characteristics and initial tumor biopsy samples of 31 inoperable LA-NSCLC patients treated with concurrent CRT. Prognostic impact of tumor cell PD-L1 expression (0% versus ≥1%) and CD8+ TIL density (0–40% vs. 41–100%) for local control, progression-free (PFS) and overall survival (OS) as well as correlations with clinicopathological features were evaluated. Results Median OS was 14 months (range: 3–167 months). The OS rates at 1- and 2 years were 68 and 20%. Local control of the entire cohort at 1 and 2 years were 74 and 61%. Median PFS, 1-year and 2-year PFS were 13 ± 1.4 months, 58 and 19%. PD-L1 expression < 1% on tumor cells was associated with improved OS, PFS and local control in patients treated with concurrent CRT. Univariate analysis showed a trend towards improved OS and local control in patients with low CD8+ TIL density. Evaluation of Tumor Immunity in the MicroEnvironment (TIME) appears to be an independent prognostic factor for local control, PFS and OS. The longest and shortest OS were achieved in patients with type I (PD-L1neg/CD8low) and type IV (PD-L1pos/CD8low) tumors (median OS: 57 ± 37 vs. 10 ± 5 months, p = 0.05), respectively. Conclusion Assessment of PD-L1 expression on tumor cells in combination with CD8+ TIL density can be a predictive biomarker in patients with inoperable LA-NSCLC treated with concurrent CRT.
Collapse
Affiliation(s)
- Kathrin Gennen
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany. .,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany. .,German Cancer Consortium (DKTK), partner site Munich, Munich, Germany.
| | - Julian Taugner
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Monika Karin
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Olarn Roengvoraphoj
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Jens Neumann
- Institute of Pathology, Faculty of Medicine, LMU, Munich, Germany
| | - Amanda Tufman
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Internal Medicine V, Thoracic Oncology Centre Munich, Division of Respiratory Medicine and Thoracic Oncology, LMU, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Simone Reu
- Department of Pathology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany.,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany.,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| |
Collapse
|
118
|
Zhou K, Guo S, Li F, Sun Q, Liang G. Exosomal PD-L1: New Insights Into Tumor Immune Escape Mechanisms and Therapeutic Strategies. Front Cell Dev Biol 2020; 8:569219. [PMID: 33178688 PMCID: PMC7593554 DOI: 10.3389/fcell.2020.569219] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
As a classical immune checkpoint molecule, PD-L1 on the surface of tumor cells plays a pivotal role in tumor immunosuppression, primarily by inhibiting the antitumor activities of T cells by binding to its receptor PD-1. PD-1/PD-L1 inhibitors have demonstrated unprecedented promise in treating various human cancers with impressive efficacy. However, a significant portion of cancer patients remains less responsive. Therefore, a better understanding of PD-L1-mediated immune escape is imperative. PD-L1 can be expressed on the surface of tumor cells, but it is also found to exist in extracellular forms, such as on exosomes. Recent studies have revealed the importance of exosomal PD-L1 (ExoPD-L1). As an alternative to membrane-bound PD-L1, ExoPD-L1 produced by tumor cells also plays an important regulatory role in the antitumor immune response. We review the recent remarkable findings on the biological functions of ExoPD-L1, including the inhibition of lymphocyte activities, migration to PD-L1-negative tumor cells and immune cells, induction of both local and systemic immunosuppression, and promotion of tumor growth. We also discuss the potential implications of ExoPD-L1 as a predictor for disease progression and treatment response, sensitive methods for detection of circulating ExoPD-L1, and the novel therapeutic strategies combining the inhibition of exosome biogenesis with PD-L1 blockade in the clinic.
Collapse
Affiliation(s)
- Kaijian Zhou
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Shu Guo,
| | - Fei Li
- Department of Pharmaceutical Science, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qiang Sun
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guoxin Liang
- Cancer Therapy Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
119
|
Niu B, Appleby TC, Wang R, Morar M, Voight J, Villaseñor AG, Clancy S, Wise S, Belzile JP, Papalia G, Wong M, Brendza KM, Lad L, Gross ML. Protein Footprinting and X-ray Crystallography Reveal the Interaction of PD-L1 and a Macrocyclic Peptide. Biochemistry 2019; 59:541-551. [PMID: 31841311 DOI: 10.1021/acs.biochem.9b00822] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Blocking interactions between PD-1 and PD-L1 opens a new era of cancer treatment involving immunity modulation. Although most immunotherapies use monoclonal antibodies, small-molecule inhibitors offer advantages. To facilitate development of small-molecule therapeutics, we implemented a rapid approach to characterize the binding interfaces of small-molecule inhibitors with PD-L1. We determined its interaction with a synthetic macrocyclic peptide by using two mass spectrometry-based approaches, hydrogen-deuterium exchange and fast photochemical oxidation of proteins (FPOP), and corroborated the findings with our X-ray structure of the PD-L1/macrocycle complex. Although all three approaches show that the macrocycle binds directly to PD-L1 over the regions of residues 46-87 and 114-125, the two protein footprinting approaches show additional binding at the N-terminus of PD-L1, and FPOP reveals some critical binding residues. The outcomes not only show the binding regions but also demonstrate the utility of MS-based footprinting in probing protein/ligand inhibitory interactions in cancer immunotherapy.
Collapse
Affiliation(s)
- Ben Niu
- Department of Chemistry , Washington University in St. Louis , St. Louis , Missouri 63130 , United States
| | - Todd C Appleby
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Ruth Wang
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Mariya Morar
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Johannes Voight
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Armando G Villaseñor
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Sheila Clancy
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Sarah Wise
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Jean-Philippe Belzile
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Giuseppe Papalia
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Melanie Wong
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Katherine M Brendza
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Latesh Lad
- Gilead Sciences, Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Michael L Gross
- Department of Chemistry , Washington University in St. Louis , St. Louis , Missouri 63130 , United States
| |
Collapse
|
120
|
|
121
|
El-Haj M, Kanovitch D, Ilan Y. Personalized inherent randomness of the immune system is manifested by an individualized response to immune triggers and immunomodulatory therapies: a novel platform for designing personalized immunotherapies. Immunol Res 2019; 67:337-347. [DOI: 10.1007/s12026-019-09101-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
122
|
Bridges JF, la Cruz MD, Pavilack M, Flood E, Janssen EM, Chehab N, Fernandes AW. Patient preferences for attributes of tyrosine kinase inhibitor treatments for EGFR mutation-positive non-small-cell lung cancer. Future Oncol 2019; 15:3895-3907. [PMID: 31621403 DOI: 10.2217/fon-2019-0396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: EGFR-tyrosine kinase inhibitors (TKIs) vary in efficacy, side effects (SEs) and dosing regimen. We explored EGFR-TKI treatment attribute preferences in EGFR mutation-positive metastatic non-small-cell lung cancer. Materials & methods: Patients completed a survey utilizing preference elicitation methods: direct elicitation of four EGFR-TKI profiles describing progression-free survival (PFS), severe SE risk, administration; discrete choice experiment involving 12 choice tasks. Results: 90 participated. The preferred profile (selected 89% of times) had the longest PFS (18 months) and the lowest severe SE risk (5%). Patients would need compensation with ≥three-times longer PFS for severe SEs. Patients would accept ≤7 months PFS reduction for oral treatments versus intravenous. Conclusion: Patients preferred longer PFS but were willing to accept reduced PFS for more favorable SEs and dosing convenience.
Collapse
Affiliation(s)
- John Fp Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | | | | | - Emuella Flood
- Patient Centered Outcomes, ICON, Gaithersburg, MD 20807, USA.,AstraZeneca, Gaithersburg, MD 20807, USA
| | - Ellen M Janssen
- Patient Centered Outcomes, ICON, Gaithersburg, MD 20807, USA
| | | | | |
Collapse
|
123
|
Cui J, Li W, Liu G, Chen X, Gao X, Lu H, Lin D. A novel circular RNA, hsa_circ_0043278, acts as a potential biomarker and promotes non-small cell lung cancer cell proliferation and migration by regulating miR-520f. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:810-821. [PMID: 30873868 DOI: 10.1080/21691401.2019.1575847] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
More and more circular RNAs (circRNAs) revealed to play a critical role in the initiation and progression of cancer, however, the effects of circRNAs on non-small cell lung cancer (NSCLC) remain largely undetermined. In the present study, we screened the dysregulated circRNAs in paired NSCLC and normal samples from GEO database and identified circ_0043278 was to be significantly up-regulated in NSCLC and demonstrated it promotes NSCLC progression in vitro and in vivo. Then, we revealed the expression of miR-520f, a downstream factor of circ_0043278, was significantly down-regulated in NSCLC and acted as a tumor inhibitor. In addition, we revealed that circ_0043278 sponged miR-520f, which was demonstrated to target ROCK1, CDKN1B, and AKT3 in NSCLC cells. In conclusion, circ_0043278 promoted NSCLC cell proliferation, invasion, and migration by increasing ROCK1, CDKN1B, and AKT3 expressions through direct inhibition of miR-520f.
Collapse
Affiliation(s)
- Jinggang Cui
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| | - Wei Li
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| | - Guohua Liu
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| | - Xiaojun Chen
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| | - Xiaolai Gao
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| | - Huiling Lu
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| | - Dang Lin
- a Department of Respiratory Medicine , Suzhou Municipal Hospital, Suzhou Hospital Affiliated to Nanjing Medical University , Suzhou , China
| |
Collapse
|
124
|
The role of DNA-demethylating agents in cancer therapy. Pharmacol Ther 2019; 205:107416. [PMID: 31626871 DOI: 10.1016/j.pharmthera.2019.107416] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/20/2019] [Indexed: 12/29/2022]
Abstract
DNA methylation patterns are frequently altered in cancer cells as compared to normal cells. A large body of research associates these DNA methylation aberrations with cancer initiation and progression. Moreover, cancer cells seem to depend upon these aberrant DNA methylation profiles to thrive. Finally, DNA methylation modifications are reversible, highlighting the potential to target the global methylation patterns for cancer therapy. In this review, we will discuss the scientific and clinical aspects of DNA methylation in cancer. We will review the limited success of targeting DNA methylation in the clinic, the associated clinical challenges, the impact of novel DNA methylation inhibitors and how combination therapies are improving patient outcomes.
Collapse
|
125
|
Li N, Zhan X. Mitochondrial Dysfunction Pathway Networks and Mitochondrial Dynamics in the Pathogenesis of Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:690. [PMID: 31649621 PMCID: PMC6794370 DOI: 10.3389/fendo.2019.00690] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Mitochondrion is a multi-functional organelle, which is associated with various signaling pathway networks, including energy metabolism, oxidative stress, cell apoptosis, cell cycles, autophagy, and immunity process. Mitochondrial proteins have been discovered to modulate these signaling pathway networks, and multiple biological behaviors to adapt to various internal environments or signaling events of human pathogenesis. Accordingly, mitochondrial dysfunction that alters the bioenergetic and biosynthetic state might contribute to multiple diseases, including cell transformation and tumor. Multiomics studies have revealed that mitochondrial dysfunction, oxidative stress, and cell cycle dysregulation signaling pathways operate in human pituitary adenomas, which suggest mitochondria play critical roles in pituitary adenomas. Some drugs targeting mitochondria are found as a therapeutic strategy for pituitary adenomas, including melatonin, melatonin inhibitors, temozolomide, pyrimethamine, 18 beta-glycyrrhetinic acid, gossypol acetate, Yougui pill, T-2 toxin, grifolic acid, cyclosporine A, dopamine agonists, and paeoniflorin. This article reviews the latest experimental evidence and potential biological roles of mitochondrial dysfunction and mitochondrial dynamics in pituitary adenoma progression, potential molecular mechanisms between mitochondria and pituitary adenoma progression, and current status and perspectives of mitochondria-based biomarkers and targeted drugs for effective management of pituitary adenomas.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
126
|
Incorvaia L, Fanale D, Badalamenti G, Barraco N, Bono M, Corsini LR, Galvano A, Gristina V, Listì A, Vieni S, Gori S, Bazan V, Russo A. Programmed Death Ligand 1 (PD-L1) as a Predictive Biomarker for Pembrolizumab Therapy in Patients with Advanced Non-Small-Cell Lung Cancer (NSCLC). Adv Ther 2019; 36:2600-2617. [PMID: 31432460 PMCID: PMC6822831 DOI: 10.1007/s12325-019-01057-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Indexed: 12/11/2022]
Abstract
Recently, immunotherapy has been shown to be an effective and helpful therapeutic option for the treatment of advanced non-small-cell lung cancer (NSCLC). The activity of antitumor T cells may be restored through the checkpoint blockade using anti-programmed death 1 or anti-programmed death ligand 1 (PD-L1) antibodies, showing, in several cancer patients, an increased progression-free survival and overall survival compared with classical chemotherapy. As recently shown by several studies, the PD-L1 expression levels in tumors may offer a selection criterion for patients to predict their immunotherapy response. In particular, NSCLC patients with high tumor PD-L1 levels (proportional score ≥ 50% for first-line therapy and ≥ 1% for second-line treatment, respectively) showed better response rates to immunotherapy and longer survival in first-line therapy compared with conventional chemotherapy. PD-L1, whose expression is evaluated by using immunohistochemistry analysis, is currently the only biomarker approved for clinical use in the first- and second-line monotherapy setting and therefore plays a central role in treatment decision-making for patients with advanced NSCLC. In this review we will discuss the key role of PD-L1 as a predictive biomarker of response to pembrolizumab therapy in NSCLC patients by describing the appropriate techniques and methodologies for immunohistochemical evaluation of PD-L1 expression and providing an overview of the clinical studies supporting its predictive significance.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Marco Bono
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Valerio Gristina
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Angela Listì
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Salvatore Vieni
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Stefania Gori
- Department of Oncology, IRCCS Ospedale Sacro Cuore-Don Calabria, Negrar, Verona, Italy
- Italian Association of Medical Oncology (AIOM), Bergamo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy.
| |
Collapse
|
127
|
Chrysin enhances anticancer drug-induced toxicity mediated by the reduction of claudin-1 and 11 expression in a spheroid culture model of lung squamous cell carcinoma cells. Sci Rep 2019; 9:13753. [PMID: 31551535 PMCID: PMC6760125 DOI: 10.1038/s41598-019-50276-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022] Open
Abstract
The aberrant expression of claudins (CLDNs), which are tight junctional proteins, is seen in various solid tumors, but the regulatory mechanisms and their pathophysiological role are not well understood. Both CLDN1 and CLDN11 were highly expressed in human lung squamous cell carcinoma (SCC). Chrysin, found in high concentration in honey and propolis, decreased CLDN1 and CLDN11 expression in RERF-LC-AI cells derived from human lung SCC. The phosphorylation level of Akt was decreased by chrysin, but those of ERK1/2 and c-Jun were not. LY-294002, an inhibitor of phosphatidylinositol 3-kinase, inhibited the phosphorylation of Akt and decreased the expression levels of CLDN1 and CLDN11. The association between phosphoinositide-dependent kinase 1 (PDK1) and Akt was inhibited by chrysin, but the phosphorylation of PDK1 was not. Immunoprecipitation and quartz-crystal microbalance assays revealed that biotinylated-chrysin binds directly to Akt. The knockdown of CLDN1 and CLDN11 using small interfering RNAs increased the transepithelial flux of doxorubicin (DXR), an anthracycline anticancer drug. Similarly, both chrysin and LY-294002 increased DXR flux. Neither CLDN1 knockdown, CLDN11 knockdown, nor chrysin changed the anticancer drug-induced cytotoxicity in a two-dimensional culture model, whereas they enhanced cytotoxicity in a spheroid culture model. Taken together, chrysin may bind to Akt and inhibit its phosphorylation, resulting in the elevation of anticancer drug-induced toxicity mediated by reductions in CLDN1 and CLDN11 expression in RERF-LC-AI cells. We suggest that chrysin may be useful as an adjuvant chemotherapy in lung SCC.
Collapse
|
128
|
Luo F, Luo M, Rong QX, Zhang H, Chen Z, Wang F, Zhao HY, Fu LW. Niclosamide, an antihelmintic drug, enhances efficacy of PD-1/PD-L1 immune checkpoint blockade in non-small cell lung cancer. J Immunother Cancer 2019; 7:245. [PMID: 31511071 PMCID: PMC6739982 DOI: 10.1186/s40425-019-0733-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022] Open
Abstract
Background PD-1/PD-L1 blockade has received approval for clinical application due to its encouraging benefit with improving prognosis in selected populations. Unfortunately, the response to immunotherapy for many patients remains unsatisfactory. It remains a great challenge to generate potential combinations that will outperform single agents alone with regard to anti-tumor activity. Methods Using NSCLC cell lines and mouse models, we explored the effects of combined niclosamide and PD-L1 blockade on tumor growth and T cell function. Furthermore, we investigated the relationship between PD-L1 and p-STAT3 expression in tumor samples from patients with NSCLC using IHC, as well as their relationship to patient survival. Results In vitro, niclosamide, an antihelmintic drug, enhanced the cancer cell lysis mediated by T cells in the presence of PD-L1 blockade. Accordingly, mice treated with niclosamide and PD-L1 antibody showed significant delay in tumor growth and increased survival which were associated with the increase of tumor infiltrating T cells and granzyme B release. Importantly, we found niclosamide could decrease the expression of PD-L1 in both a concentration- and time-dependent manner in NSCLC cells, which was linked to the blockage of p-STAT3 binding to the promoter of PD-L1. Conclusions An enhancement of PD-L1 antibody by niclosamide was observed in inhibition of NSCLC growth in vitro and in vivo, which was involved in blockage of p-STAT3 binding to promoter of PD-L1 and finally downregulation of PD-L1 expression. These encourage the combination therapy of niclosamide and PD-1/PD-L1 blockade to be further studied in clinic. Supplementary information Supplementary information accompanies this paper at 10.1186/s40425-019-0733-7.
Collapse
Affiliation(s)
- Fan Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Min Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Qi-Xiang Rong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhen Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hong-Yun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Li-Wu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
129
|
Molecular basis and rationale for combining immune checkpoint inhibitors with chemotherapy in non-small cell lung cancer. Drug Resist Updat 2019; 46:100644. [PMID: 31585395 DOI: 10.1016/j.drup.2019.100644] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022]
|
130
|
Wu CH, Hwang MJ. Risk stratification for lung adenocarcinoma on EGFR and TP53 mutation status, chemotherapy, and PD-L1 immunotherapy. Cancer Med 2019; 8:5850-5861. [PMID: 31407494 PMCID: PMC6792489 DOI: 10.1002/cam4.2492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022] Open
Abstract
The overall survival rates for lung cancer remain unsatisfactorily low, even for patients with biomarkers for which target therapies or immunotherapies are recommended. Better identification of at‐risk patients is needed to achieve more effective personalized treatment. Here, we derived a risk‐stratifying gene signature consisting of five genes that had the greatest differential expression by stage from lung adenocarcinoma (LUAD) transcriptomes. The new gene signature enabled survival prognosis for multiple LUAD datasets from different platforms of transcriptomics and risk stratification for patients with and without a mutation in TP53 or EGFR, with high and low levels of PD‐L1, and with and without adjuvant chemotherapy treatment. Using these evaluations, it was also shown to be more robust compared to several other gene signatures. Functional analysis of the five genes and their protein‐protein interaction partners indicated that they are functionally enriched in cell cycle, endocytosis, and EGFR regulation, which are biological processes associated with lung cancer and drug resistance. Extensive discussions on related experimental studies suggest that the five genes are novel and sensible targets for developing new drugs and/or tackling drug resistance problems for LUAD.
Collapse
Affiliation(s)
- Chih-Hsun Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ming-Jing Hwang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
131
|
Valentini AM, Di Pinto F, Coletta S, Guerra V, Armentano R, Caruso ML. Tumor microenvironment immune types in gastric cancer are associated with mismatch repair however, not HER2 status. Oncol Lett 2019; 18:1775-1785. [PMID: 31423245 PMCID: PMC6614673 DOI: 10.3892/ol.2019.10513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
The treatment of patients with human epidermal growth factor receptor 2 (HER2)-negative gastric cancer is a major challenge. Immunotherapy using immune checkpoint inhibitors is a rapidly growing field. In a number of malignancy types it has been demonstrated that patients with mismatch repair deficiency efficiently respond to programmed death-ligand 1 (PD-L1) blockade therapy. Recent studies have evaluated tumor microenvironment immune types to predict which patients may clinically benefit from immunotherapy. The present study aimed to evaluate the immunohistochemical expression of PD-L1 in 70 gastric cancer tissue samples. Potential associations between PD-L1 expression and mismatch repair deficiency, HER2 and Epstein Barr virus (EBV) status were then investigated in the context of the tumor microenvironment. A positive association was identified for PD-L1 expression with mismatch repair deficiency and EBV status; however, no association was revealed with HER2 status. Immunohistochemistry was then used to classify the microenvironment immune types. This demonstrated that the majority of the gastric cancer samples (73%) belonged to the tumor microenvironment immune type II [PD-L1-/cluster of differentiation 8 (CD8)+ low], which involves an immune ignorant state and has a low sensitivity to immunotherapy. However, 7% of the gastric cancer cases were identified to belong to the tumor microenvironment immune type I (PD-L1+/CD8+ high), which exhibits adaptive immune escape responses and a high chance of reversion with immune checkpoint blockade therapy. In conclusion, the present study emphasized the importance of evaluating tumor microenvironment immune types, mismatch repair deficiency status and EBV status, rather than PD-L1 expression alone, when evaluating the eligibility of a patient for immunotherapy with anti-programmed cell death protein-1/PD-L1 antibodies.
Collapse
Affiliation(s)
- Anna Maria Valentini
- Department of Pathology, National Institute of Gastroenterology ‘S. de Bellis’, Research Hospital, Castellana Grotte, I-70013 Bari, Italy
| | - Federica Di Pinto
- Department of Pathology, National Institute of Gastroenterology ‘S. de Bellis’, Research Hospital, Castellana Grotte, I-70013 Bari, Italy
| | - Sergio Coletta
- Department of Pathology, National Institute of Gastroenterology ‘S. de Bellis’, Research Hospital, Castellana Grotte, I-70013 Bari, Italy
| | - Vito Guerra
- Department of Epidemiology, National Institute of Gastroenterology ‘S. de Bellis’, Research Hospital, Castellana Grotte, I-70013 Bari, Italy
| | - Raffaele Armentano
- Department of Pathology, National Institute of Gastroenterology ‘S. de Bellis’, Research Hospital, Castellana Grotte, I-70013 Bari, Italy
| | - Maria Lucia Caruso
- Department of Pathology, National Institute of Gastroenterology ‘S. de Bellis’, Research Hospital, Castellana Grotte, I-70013 Bari, Italy
| |
Collapse
|
132
|
Shen T, Chen Z, Qiao J, Sun X, Xiao Q. Neutralizing monoclonal antibody against Dickkopf2 impairs lung cancer progression via activating NK cells. Cell Death Discov 2019; 5:123. [PMID: 31372243 PMCID: PMC6668384 DOI: 10.1038/s41420-019-0204-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/03/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022] Open
Abstract
Adenomatous polyposis coli (APC) and KRAS proto-oncogene (KRAS) mutations frequently co-occur in non-small cell lung cancer. Inactivating APC mutations in colorectal carcinoma has been well characterized, leading to the approaches targeting on dysregulated APC pathway. However, it remains undetermined whether such approaches are also applicable to non-small cell lung cancer patients harboring similar mutations of APC. Dickkopf-related protein 2 (DKK2) is a Wnt antagonist. Our previous study has proved that anti-DKK2 antibody 5F8 suppressed the growth of colorectal carcinoma with APC mutations, illustrating a new target agent of APC-mutated tumors. This study aimed to investigate the potential of applying anti-DKK2 antibody to non-small cell lung cancer with APC mutations. We found significant upregulation of Dkk2 expression in APC-mutated lung cancers. Administration of DKK2 antibody inhibited cancer growth via modulating tumor immune microenvironment in lung cancer mouse models. Our study provided strong evidence supporting APC mutations-directed applications of anti-DKK2 targeted therapy in a wide range of cancer types, including lung cancer.
Collapse
Affiliation(s)
- Tianli Shen
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
- Department of Pharmacology, School of Medicine, Yale University, 10 Amistad St, New Haven, CT USA
| | - Zhengxi Chen
- Department of Pharmacology, School of Medicine, Yale University, 10 Amistad St, New Haven, CT USA
- Department of Orthodontics, Shanghai Ninth People׳s Hospital, School of Stomatology, Shanghai key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Ju Qiao
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA USA
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Qian Xiao
- Department of Pharmacology, School of Medicine, Yale University, 10 Amistad St, New Haven, CT USA
| |
Collapse
|
133
|
Frazer IH, Chandra J. Immunotherapy for HPV associated cancer. PAPILLOMAVIRUS RESEARCH 2019; 8:100176. [PMID: 31310819 PMCID: PMC6639647 DOI: 10.1016/j.pvr.2019.100176] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 02/01/2023]
Affiliation(s)
- Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, 4102, Australia.
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
134
|
Differential expression of individual transcript variants of PD-1 and PD-L2 genes on Th-1/Th-2 status is guaranteed for prognosis prediction in PCNSL. Sci Rep 2019; 9:10004. [PMID: 31292525 PMCID: PMC6620277 DOI: 10.1038/s41598-019-46473-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/01/2019] [Indexed: 12/22/2022] Open
Abstract
In current molecular medicine, next-generation sequencing (NGS) for transcript variant detection and multivariable analyses are valid methods for evaluating gene expression, cancer mechanisms, and prognoses of patients. We conducted RNA-sequencing on samples from patients with primary central nervous system lymphoma (PCNSL) using NGS and performed multivariable analysis on gene expression data and correlations focused on Th-1/Th-2 helper T cell balance and immune checkpoint to identify diagnosis/prognosis markers and cancer immune pathways in PCNSL. We selected 84 transcript variants to limit the analysis range for Th-1/Th-2 balance and stimulatory and inhibitory checkpoints in 31 PCNSLs. Of these, 21 highly-expressed transcript variants were composed of the formulas for prognoses based on Th-1/Th-2 status and checkpoint activities. Using formulas, Th-1low, Th-2high, and stimulatory checkpointhigh resulted in poor prognoses. Further, Th-1highTh-2low was associated with good prognoses. On the other hand, CD40-001high and CD70-001high as stimulatory genes, and LAG3-001high, PDCD1 (PD-1)-001/002/003high, and PDCD1LG2 (PD-L2)-201low as inhibitory genes were associated with poor prognoses. Interestingly, Th-1highTh-2low and Th-1lowTh-2high were correlated with stimulatory checkpointlow as CD70-001low and inhibitory checkpointlow as HAVCR2 (TIM-3)-001low and PDCD1LG2-001/201low, respectively. Focused on the inhibitory checkpoint, specific variants of CD274 (PD-L1)-001 and PDCD1-002 served severe hazard ratios. In particular, PDCD1-002high by a cut off score was associated with poor prognoses, in addition to PDCD1-001/003high, PDCD1LG2-201low, and LAG3-001high. These results mainly suggest that expression of transcript variants of PDCD1 and PDCD1LG2 on the Th-1/Th-2 balance enable prognostic prediction in PCNSL. This study provides insights for development of molecular target therapies and identification of diagnosis/prognosis markers in PCNSL.
Collapse
|
135
|
Liu SY, Huang WC, Yeh HI, Ko CC, Shieh HR, Hung CL, Chen TY, Chen YJ. Sequential Blockade of PD-1 and PD-L1 Causes Fulminant Cardiotoxicity-From Case Report to Mouse Model Validation. Cancers (Basel) 2019; 11:cancers11040580. [PMID: 31022941 PMCID: PMC6521128 DOI: 10.3390/cancers11040580] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/25/2022] Open
Abstract
The combined administration of programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors might be considered as a treatment for poorly responsive cancer. We report a patient with brain metastatic lung adenocarcinoma in whom fatal myocarditis developed after sequential use of PD-1 and PD-L1 inhibitors. This finding was validated in syngeneic tumor-bearing mice. The mice bearing lung metastases of CT26 colon cancer cells treated with PD-1 and/or PD-L1 inhibitors showed that the combination of anti-PD-1 and anti-PD-L1, either sequentially or simultaneously administered, caused myocarditis lesions with myocyte injury and patchy mononuclear infiltrates in the myocardium. A significant increase of infiltrating neutrophils in myocytes was noted only in mice with sequential blockade, implying a role for the pathogenesis of myocarditis. Among circulating leukocytes, concurrent and subsequent treatment of PD-1 and PD-L1 inhibitors led to sustained suppression of neutrophils. Among tumor-infiltrating leukocytes, combinatorial blockade increased CD8+ T cells and NKG2D+ T cells, and reduced tumor-associated macrophages, neutrophils, and natural killer (NK) cells in the lung metastatic microenvironment. The combinatorial treatments exhibited better control and anti-PD-L1 followed by anti-PD-1 was the most effective. In conclusion, the combinatory use of PD-1 and PD-L1 blockade, either sequentially or concurrently, may cause fulminant cardiotoxicity, although it gives better tumor control, and such usage should be cautionary.
Collapse
Affiliation(s)
- Shin-Yi Liu
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
| | - Wen-Chien Huang
- Department of Thoracic Surgery, MacKay Memorial Hospital, Taipei 10449, Taiwan.
| | - Hung-I Yeh
- Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.
| | - Chun-Chuan Ko
- Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.
| | - Hui-Ru Shieh
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
| | - Chung-Lieh Hung
- Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.
| | - Tung-Ying Chen
- Department of Pathology, MacKay Memorial Hospital, Taipei 10449, Taiwan.
| | - Yu-Jen Chen
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 10449, Taiwan.
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan.
| |
Collapse
|
136
|
Heim L, Kachler K, Siegmund R, Trufa DI, Mittler S, Geppert CI, Friedrich J, Rieker RJ, Sirbu H, Finotto S. Increased expression of the immunosuppressive interleukin-35 in patients with non-small cell lung cancer. Br J Cancer 2019; 120:903-912. [PMID: 30956278 PMCID: PMC6734661 DOI: 10.1038/s41416-019-0444-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/08/2019] [Accepted: 03/15/2019] [Indexed: 01/01/2023] Open
Abstract
Background The immunosuppressive role of the cytokine IL-35 in patients with non-small cell lung cancer (NSCLC) is poorly understood. In this study, we analysed the localisation and regulation of IL-35 in the lung of patients with non-small cell lung cancer (NSCLC) to further elucidate the immune-escape of cancer cells in perioperative course of disease. Methods Interleukin 35 (IL-35) was measured by ELISA in postoperative serum from 7 patients with NSCLC as well as 8 samples from healthy controls. Immunohistochemistry, FACS analysis, real-time PCR, as well as western blot from samples of the control (CTR), peri-tumoural (PT) and the tumoural (TU) region of the lung derived from patients with NSCLC and 10 controls were performed. Results Here we found elevated levels of IL-35 in the TU region as well as postoperative serum from patients with lung adenocarcinoma. Consistently, we found an increased expression of IL-35+Foxp-3+ cells, which associated with ARG1 mRNA expression and decreased TNFA in the TU region of the lung of patients with NSCLC as compared to their CTR region. Furthermore, in the CTR region of the lung of patients with NSCLC, CD68+ macrophages were induced and correlated with IL-35+ cells. Finally, IL-35 positively correlated with TTF-1+PD-L1+ cells in the TU region of NSCLC patients. Conclusions Induced IL-35+Foxp3+ cell numbers in the TU region of the lung of patients with NSCLC associated with ARG1 mRNA expression and with TTF-1+PD-L1+ cells. In the tumour-free CTR area, IL-35 correlated with CD68+ macrophages. Thus inhibitors to IL-35 would probably succeed in combination with antibodies against immune checkpoints like PD-L1 and PD-1 currently used against NSCLC because they would inhibit immunosuppressive macrophages and T regulatory cells while promoting T cell-mediated anti-tumoural immune responses in the microenvironment as well as the TU region of NSCLC patients.
Collapse
Affiliation(s)
- Lisanne Heim
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Katerina Kachler
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raphaela Siegmund
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Denis I Trufa
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Thoracic Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Susanne Mittler
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Carol-Immanuel Geppert
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Juliane Friedrich
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ralf J Rieker
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
137
|
Bozkaya Y, Yazıcı O. Prognostic significance of gamma-glutamyl transferase in patients with metastatic non-small cell lung cancer. Expert Rev Mol Diagn 2019; 19:267-272. [PMID: 30722710 DOI: 10.1080/14737159.2019.1579644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The prognostic significance of serum gamma-glutamyl transferase (GGT) level at diagnosis in patients with metastatic non-small lung cancer (NSCLC) is not clear. We aimed to assess the relationship between serum GGT level and overall survival (OS) and progression-free survival (PFS) in this patient population. METHODS Data of patients with metastatic NSCLC who were admitted to the medical oncology clinic of our hospital during April 2013-December 2017 were retrospectively analyzed. Patients were divided into two groups based on GGT levels, normal and high (as defined by normal reference levels), and then compared. RESULTS Significant differences between the high and normal GGT level groups were found regarding female sex, Eastern Cooperative Oncology Group performance score, weight loss at the time of diagnosis, and lactate dehydrogenase level (p < 0.05). The high GGT group had a shorter median OS (11.5 vs. 3.4 months, p < 0.001) and PFS (7.8 vs. 3.0 months, p = 0.001). High GGT level is an independent risk factor for OS (hazard ratio [HR] 2.270; 95% confidence interval [CI], 1.398-3.686; p < 0.001) and PFS (HR 2.489; 95% CI, 1.323-4.684; p = 0.005). CONCLUSIONS High serum GGT level is an independent prognostic factor for OS and PFS in metastatic NSCLC patients.
Collapse
Affiliation(s)
- Yakup Bozkaya
- a Clinic of Medical Oncology , Edirne State Hospital , Edirne , Turkey
| | - Ozan Yazıcı
- b Department of Medical Oncology , Gazi University Faculty of Medicine , Ankara , Turkey
| |
Collapse
|
138
|
LncRNA TP73-AS1 promoted the progression of lung adenocarcinoma via PI3K/AKT pathway. Biosci Rep 2019; 39:BSR20180999. [PMID: 30541897 PMCID: PMC6328885 DOI: 10.1042/bsr20180999] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 11/29/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022] Open
Abstract
Lung adenocarcinoma (LAD) is one of the most common malignancies that threats human health worldwide. Long non-coding RNAs (lncRNAs) have been reported to play significant roles in tumorigenesis and might be novel biomarkers and targets for diagnosis and treatment of cancers. TP73-AS1 is a newly discovered lncRNA involved in the tumorigenesis and development of several cancers. However, its role in LAD has not been investigated yet. In the present study, we first found that TP73-AS1 expression was markedly increased in LAD tissues and cell lines and its overexpression was strongly associated with poor clinical outcomes. Then the loss/gain-of-function assays elucidated that TP73-AS1 contributed to cell proliferation, migration, and invasion in vitro, and the in vivo experiments illustrated that its knockdown inhibited tumor growth and metastasis. What was more, we discovered that phosphoinositide 3-kinase and AKT (PI3K/AKT) pathway was activated both in LAD tissues and cell lines but inactivated under TP73-AS1 silence. Moreover, the activation of this pathway could rescue the inhibitory effects of TP73-AS1 suppression on LAD cellular processes partially. These data suggested that TP73-AS1 served as an oncogene in LAD partially through activating PI3K/AKT pathway and it could be a potential target for diagnosis and treatment of LAD.
Collapse
|
139
|
|
140
|
Hsu PC, Yang CT, Jablons DM, You L. The Role of Yes-Associated Protein (YAP) in Regulating Programmed Death-Ligand 1 (PD-L1) in Thoracic Cancer. Biomedicines 2018; 6:biomedicines6040114. [PMID: 30544524 PMCID: PMC6315659 DOI: 10.3390/biomedicines6040114] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022] Open
Abstract
The programmed death-ligand 1(PD-L1)/PD-1 pathway is an immunological checkpoint in cancer cells. The binding of PD-L1 and PD-1 promotes T-cell tolerance and helps tumor cells escape from host immunity. Immunotherapy targeting the PD-L1/PD-1 axis has been developed as an anti-cancer therapy and used in treating advanced human non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). Yes-associated protein (YAP) is a key mediator of the Hippo/YAP signaling pathway, and plays important roles in promoting cancer development, drug resistance and metastasis in human NSCLC and MPM. YAP has been suggested as a new therapeutic target in NSCLC and MPM. The role of YAP in regulating tumor immunity such as PD-L1 expression has just begun to be explored, and the correlation between YAP-induced tumorigenesis and host anti-tumor immune responses is not well known. Here, we review recent studies investigating the correlation between YAP and PD-L1 and demonstrating the mechanism by which YAP regulates PD-L1 expression in human NSCLC and MPM. Future work should focus on the interactions between Hippo/YAP signaling pathways and the immune checkpoint PD-L1/PD-1 pathway. The development of new synergistic drugs for immune checkpoint PD-L1/PD-1 blockade in NSCLC and MPM is warranted.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA.
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan.
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan.
| | - David M Jablons
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA.
| | - Liang You
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA.
| |
Collapse
|